WO2010030704A2 - Antibacterial aminoglycoside analogs - Google Patents

Antibacterial aminoglycoside analogs Download PDF

Info

Publication number
WO2010030704A2
WO2010030704A2 PCT/US2009/056407 US2009056407W WO2010030704A2 WO 2010030704 A2 WO2010030704 A2 WO 2010030704A2 US 2009056407 W US2009056407 W US 2009056407W WO 2010030704 A2 WO2010030704 A2 WO 2010030704A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
hydrogen
mmol
yield
reaction
Prior art date
Application number
PCT/US2009/056407
Other languages
French (fr)
Other versions
WO2010030704A3 (en
Inventor
James Bradley Aggen
Martin Sheringham Linsell
Adam Aaron Goldblum
Darin James Hildebrandt
Timothy Robert Kane
Paola Dozzo
Micah James Gliedt
Stephen Hanessian
Alexandre Giguere
Justyna Grzyb
Juan Pablo Maianti
Original Assignee
Achaogen, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Achaogen, Inc. filed Critical Achaogen, Inc.
Publication of WO2010030704A2 publication Critical patent/WO2010030704A2/en
Publication of WO2010030704A3 publication Critical patent/WO2010030704A3/en
Priority to US13/044,234 priority Critical patent/US8399419B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/20Carbocyclic rings
    • C07H15/22Cyclohexane rings, substituted by nitrogen atoms
    • C07H15/222Cyclohexane rings substituted by at least two nitrogen atoms
    • C07H15/226Cyclohexane rings substituted by at least two nitrogen atoms with at least two saccharide radicals directly attached to the cyclohexane rings
    • C07H15/228Cyclohexane rings substituted by at least two nitrogen atoms with at least two saccharide radicals directly attached to the cyclohexane rings attached to adjacent ring-carbon atoms of the cyclohexane rings
    • C07H15/232Cyclohexane rings substituted by at least two nitrogen atoms with at least two saccharide radicals directly attached to the cyclohexane rings attached to adjacent ring-carbon atoms of the cyclohexane rings with at least three saccharide radicals in the molecule, e.g. lividomycin, neomycin, paromomycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents

Definitions

  • the present invention is directed to novel aminoglycoside compounds, and methods for their preparation and use as therapeutic or prophylactic agents.
  • RNA which serves as a messenger between DNA and proteins, was thought to be an entirely flexible molecule without significant structural complexity. Recent studies have revealed a surprising intricacy in RNA structure. RNA has a structural complexity rivaling proteins, rather than simple motifs like DNA. Genome sequencing reveals both the sequences of the proteins and the mRNAs that encode them. Since proteins are synthesized using an RNA template, such proteins can be inhibited by preventing their production in the first place by interfering with the translation of the mRNA. Since both proteins and the RNAs are potential drug targeting sites, the number of targets revealed from genome sequencing efforts is effectively doubled. These observations unlock a new world of opportunities for the pharmaceutical industry to target RNA with small molecules.
  • Proteins can be extremely difficult to isolate and purify in the appropriate form for use in assays for drug screening. Many proteins require post-translational modifications that occur only in specific cell types under specific conditions. Proteins fold into globular domains with hydrophobic cores and hydrophilic and charged groups on the surface. Multiple subunits frequently form complexes, which may be required for a valid drug screen. Membrane proteins usually need to be embedded in a membrane to retain their proper shape. The smallest practical unit of a protein that can be used in drug screening is a globular domain.
  • RNAs are essentially equivalent in their solubility, ease of synthesis or use in assays.
  • the physical properties of RNAs are independent of the protein they encode. They may be readily prepared in large quantity through either chemical or enzymatic synthesis and are not extensively modified in vivo.
  • RNA the smallest practical unit for drug binding is the functional subdomain.
  • a functional subdomain in RNA is a fragment that, when removed from the larger RNA and studied in isolation, retains its biologically relevant shape and protein or RNA-binding properties. The size and composition of RNA functional subdomains make them accessible by enzymatic or chemical synthesis.
  • RNA subdomains The structural biology community has developed significant experience in identification of functional RNA subdomains in order to facilitate structural studies by techniques such as NMR spectroscopy.
  • small analogs of the decoding region of 16S rRNA the A-site have been identified as containing only the essential region, and have been shown to bind antibiotics in the same fashion as the intact ribosome.
  • RNA binding sites on RNA are hydrophilic and relatively open as compared to proteins.
  • the potential for small molecule recognition based on shape is enhanced by the deformability of RNA.
  • the binding of molecules to specific RNA targets can be determined by global conformation and the distribution of charged, aromatic, and hydrogen bonding groups off of a relatively rigid scaffold. Properly placed positive charges are believed to be important, since long-range electrostatic interactions can be used to steer molecules into a binding pocket with the proper orientation. In structures where nucleobases are exposed, stacking interactions with aromatic functional groups may contribute to the binding interaction.
  • the major groove of RNA provides many sites for specific hydrogen bonding with a ligand.
  • RNA RNA molecules
  • aromatic N7 nitrogen atoms of adenosine and guanosine the 04 and O6 oxygen atoms of undine and guanosine
  • the amines of adenosine and cytidine The rich structural and sequence diversity of RNA suggests to us that ligands can be created with high affinity and specificity for their target.
  • Certain small molecules can bind and block essential functions of RNA.
  • examples of such molecules include the aminoglycoside antibiotics and drugs such as erythromycin which binds to bacterial rRNA and releases peptidyl-tRNA and mRNA.
  • Aminoglycoside antibiotics have long been known to bind RNA. They exert their antibacterial effects by binding to specific target sites in the bacterial ribosome. For the structurally related antibiotics neamine, ribostamycin, neomycin B, and paromomycin, the binding site has been localized to the A-site of the prokaryotic 16S ribosomal decoding region RNA (see Moazed, D.; Noller, H.F., Nature, 1987, 327, 389).
  • RNA targets in the ribosome one of the most ancient and conserved targets in bacteria. Since antibacterial drugs are desired to be potent and have broad-spectrum activity, these ancient processes, fundamental to all bacterial life, represent attractive targets. The closer we get to ancient conserved functions the more likely we are to find broadly conserved RNA shapes.
  • RNA of the small ribosomal subunit The bactericidal action is mediated by binding to the bacterial RNA in a fashion that leads to misreading of the genetic code. Misreading of the code during translation of integral membrane proteins is thought to produce abnormal proteins that compromise the barrier properties of the bacterial membrane.
  • Antibiotics are chemical substances produced by various species of microorganisms (bacteria, fungi, actinomycetes) that suppress the growth of other microorganisms and may eventually destroy them.
  • antibiotics common usage often extends the term antibiotics to include synthetic antibacterial agents, such as the sulfonamides, and quinolines, that are not products of microbes.
  • the number of antibiotics that have been identified now extends into the hundreds, and many of these have been developed to the stage where they are of value in the therapy of infectious diseases.
  • Antibiotics differ markedly in physical, chemical, and pharmacological properties, antibacterial spectra, and mechanisms of action. In recent years, knowledge of molecular mechanisms of bacterial, fungal, and viral replication has greatly facilitated rational development of compounds that can interfere with the life cycles of these microorganisms.
  • resistance to antibiotics usually involves a stable genetic change, inheritable from generation to generation. Any of the mechanisms that result in alteration of bacterial genetic composition can operate. While mutation is frequently the cause, resistance to antimicrobial agents may be acquired through transfer of genetic material from one bacterium to another by transduction, transformation or conjugation.
  • the present invention is directed to novel aminoglycoside compounds, having antibacterial activity, including stereoisomers, pharmaceutically acceptable salts and prodrugs thereof, and the use of such compounds in the treatment of bacterial infections.
  • Q 2 is hydrogen, optionally substituted alkyl
  • Zi and Z 2 are both hydrogen or Zj and Z 2 form a double bond
  • Z 3 and Z 4 are both hydrogen or Z 3 and Z 4 form a double bond.
  • a pharmaceutical composition comprising a compound having structure (I), or a stereoisomer, pharmaceutically acceptable salt or prodrug thereof, and a pharmaceutically acceptable carrier, diluent or excipient.
  • the present invention provides a method of treating a bacterial infection in a mammal comprising administering to the mammal an effective amount of a compound having structure (I), or a stereoisomer, pharmaceutically acceptable salt or prodrug thereof.
  • Niro refers to the -NO 2 radical.
  • Alkyl refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds), having from one to twelve carbon atoms (Ci-Ci 2 alkyl), preferably one to eight carbon atoms (C)-Cg alkyl) or one to six carbon atoms (Ci-C 6 alkyl), and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, n-propyl, 1 -methyl ethyl (zs ⁇ -propyl), «-butyl, n-pentyl,
  • 1,1 -dimethyl ethyl (f-butyl), 3-methylhexyl, 2-methylhexyl, ethenyl, prop-1-enyl, but-1-enyl, pent-1-enyl, penta-l,4-dienyl, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like.
  • an alkyl group may be optionally substituted.
  • Alkylene or “alkylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds), and having from one to twelve carbon atoms, e.g., methylene, ethylene, propylene, rc-butylene, ethenylene, propenylene, w-butenylene, propynylene, n-butynylene, and the like.
  • the alkylene chain is attached to the rest of the molecule through a single or double bond and to the radical group through a single or double bond.
  • the points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, an alkylene chain may be optionally substituted.
  • Alkoxy refers to a radical of the formula -OR a where R 3 is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkoxy group may be optionally substituted.
  • Alkylamino refers to a radical of the formula -NHR a or -NR 3 R 3 where each R 3 is, independently, an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkylamino group may be optionally substituted.
  • Thioalkyl refers to a radical of the formula -SR 3 where R 3 is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, a thioalkyl group may be optionally substituted.
  • Aryl refers to a hydrocarbon ring system radical comprising hydrogen
  • the aryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems.
  • Aryl radicals include, but are not limited to, aryl radicals derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, ⁇ zs-indacene, s-indacene, indane, indene, naphthalene, phenalene, phenanthrene, pleiadene, pyrene, and triphenylene.
  • Aralkyl refers to a radical of the formula -R b -R c where R b is an alkylene chain as defined above and R c is one or more aryl radicals as defined above, for example, benzyl, diphenylmethyl and the like. Unless stated otherwise specifically in the specification, an aralkyl group may be optionally substituted.
  • Cycloalkyl or “carbocyclic ring” refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which may include fused or bridged ring systems, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond.
  • Monocyclic radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • Polycyclic radicals include, for example, adamantyl, norbornyl, decalinyl, 7,7-dimethyl-bicyclo[2.2.1]heptanyl, and the like. Unless otherwise stated specifically in the specification, a cycloalkyl group may be optionally substituted.
  • Cycloalkylalkyl refers to a radical of the formula -R b Ra where Ra is an alkylene chain as defined above and R g is a cycloalkyl radical as defined above. Unless stated otherwise specifically in the specification, a cycloalkylalkyl group may be optionally substituted.
  • fused refers to any ring structure described herein which is fused to an existing ring structure in the compounds of the invention.
  • the fused ring is a heterocyclyl ring or a heteroaryl ring
  • any carbon atom on the existing ring structure which becomes part of the fused heterocyclyl ring or the fused heteroaryl ring may be replaced with a nitrogen atom.
  • Halo or halogen refers to bromo, chloro, fluoro or iodo.
  • Haloalkyl refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1,2-difluoroethyl,
  • haloalkyl group may be optionally substituted.
  • Heterocyclyl or “heterocyclic ring” refers to a stable 3- to 18-membered non-aromatic ring radical which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur.
  • the heterocyclyl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized; and the heterocyclyl radical may be partially or fully saturated.
  • heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl[l,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, mo ⁇ holinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1 -oxox
  • 'W-heterocyclyl refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a nitrogen atom in the heterocyclyl radical. Unless stated otherwise specifically in the specification, a 7V-heterocyclyl group may be optionally substituted.
  • Heterocyclylalkyl refers to a radical of the formula -R b R e where R b is an alkylene chain as defined above and R e is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl may be attached to the alkyl radical at the nitrogen atom. Unless stated otherwise specifically in the specification, a heterocyclylalkyl group may be optionally substituted.
  • Heteroaryl refers to a 5- to 14-membered ring system radical comprising hydrogen atoms, one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and at least one aromatic ring.
  • the heteroaryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized.
  • Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzothiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[ ⁇ ][l,4]dioxepinyl, 1 ,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[l,2-a]pyridinyl, carbazolyl, cinnolinyl, dibenzofuranyl, dibenzothiopheny
  • a heteroaryl group may be optionally substituted.
  • 'W-heteroaryl refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical.
  • an 7V-heteroaryl group may be optionally substituted.
  • ⁇ eteroarylalkyl refers to a radical of the formula -R b R f where R b is an alkylene chain as defined above and R f is a heteroaryl radical as defined above. Unless stated otherwise specifically in the specification, a heteroarylalkyl group may be optionally substituted.
  • substituted means any of the above groups (i.e., alkyl, alkylene, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, 7V-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl) wherein at least one hydrogen atom is replaced by a bond to a non-hydrogen atoms such as, but not limited to: a halogen atom such as F, Cl, Br, and I; an oxygen atom in groups such as hydroxyl groups, alkoxy groups, and ester groups; a sulfur atom in groups such as thiol groups, thioalkyl groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such
  • Substituted also means any of the above groups in which one or more hydrogen atoms are replaced by a higher-order bond (e.g., a double- or triple-bond) to a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and ester groups; and nitrogen in groups such as imines, oximes, hydrazones, and nitriles.
  • a higher-order bond e.g., a double- or triple-bond
  • nitrogen in groups such as imines, oximes, hydrazones, and nitriles.
  • R g and R h are the same or different and independently hydrogen, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, JV-heterocyclyl, heterocyclylalkyl, heteroaryl, 7V-heteroaryl and/or heteroarylalkyl.
  • Substituted further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to an amino, cyano, hydroxyl, imino, nitro, oxo, thioxo, halo, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, 7V-heterocyclyl, heterocyclylalkyl, heteroaryl, 7V-heteroaryl and/or heteroarylalkyl group.
  • each of the foregoing substituents may also be optionally substituted with one or more of the above substituents.
  • protecting group refers to a labile chemical moiety which is known in the art to protect reactive groups including without limitation, hydroxyl and amino groups, against undesired reactions during synthetic procedures. Hydroxyl and amino groups which protected with a protecting group are referred to herein as “protected hydroxyl groups” and “protected amino groups”, respectively. Protecting groups are typically used selectively and/or orthogonally to protect sites during reactions at other reactive sites and can then be removed to leave the unprotected group as is or available for further reactions. Protecting groups as known in the art are described generally in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, New York (1999).
  • Groups can be selectively incorporated into aminoglycosides of the invention as precursors.
  • an amino group can be placed into a compound of the invention as an azido group that can be chemically converted to the amino group at a desired point in the synthesis.
  • groups are protected or present as a precursor that will be inert to reactions that modify other areas of the parent molecule for conversion into their final groups at an appropriate time. Further representative protecting or precursor groups are discussed in Agrawal, et al., Protocols for Oligonucleotide Conjugates, Eds, Humana Press; New Jersey, 1994; Vol. 26 pp. 1 -72.
  • hydroxyl protecting groups include, but are not limited to, t-butyl, t-butoxymethyl, methoxymethyl, tetrahydropyranyl, 1-ethoxyethyl, l-(2- chloroethoxy)ethyl, 2-trimethylsilylethyl, p-chlorophenyl, 2,4-dinitrophenyl, benzyl, 2,6-dichlorobenzyl, diphenylmethyl, p-nitrobenzyl, triphenylmethyl, trimethylsilyl, triethylsilyl, t-butyldimethylsilyl, t-butyldiphenylsilyl, triphenylsilyl, benzoylformate, acetate, chloroacetate, trichloroacetate, trifluoroacetate, pivaloate, benzoate, p- phenylbenzoate, 9-fluorenylmethyl carbonate, mesylate and tos
  • amino protecting groups include, but are not limited to, carbamate-protecting groups, such as 2-trimethylsilylethoxycarbonyl (Teoc), 1 -methyl- 1 -(4-biphenyl yl)ethoxy- carbonyl (Bpoc), t-butoxycarbonyl (BOC), allyloxycarbonyl (Alloc), 9- fluorenylmethyloxycarbonyl (Fmoc), and benzyloxycarbonyl (Cbz); amide protecting groups, such as formyl, acetyl, trihaloacetyl, benzoyl, and nitrophenylacetyl; sulfonamide-protecting groups, such as 2-nitrobenzenesulfonyl; and imine and cyclic imide protecting groups, such as phthalimido and dithiasuccinoyl.
  • carbamate-protecting groups such as 2-trimethylsilylethoxycarbonyl (Teoc), 1 -methyl- 1
  • Prodrug is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound of the invention.
  • prodrug refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable.
  • a prodrug may be inactive when administered to a subject in need thereof, but is converted in vivo to an active compound of the invention.
  • Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood.
  • the prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam)).
  • prodrugs of a compound of the invention may be prepared by modifying functional groups present in the compound of the invention in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound of the invention.
  • Prodrugs include compounds of the invention wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the compound of the invention is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively.
  • Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol or amide derivatives of amine functional groups in the compounds of the invention and the like.
  • the invention disclosed herein is also meant to encompass all pharmaceutically acceptable compounds of structure (I) being isotopically-labelled by having one or more atoms replaced by an atom having a different atomic mass or mass number.
  • isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2 H, 3 H, 1 1 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 0, 31 P, 32 P, 35 S, 18 F, 36 Cl, 123 I, and 125 I, respectively.
  • radiolabeled compounds could be useful to help determine or measure the effectiveness of the compounds, by characterizing, for example, the site or mode of action, or binding affinity to pharmacologically important site of action.
  • Certain isotopically-labelled compounds of structure (I) for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies.
  • the radioactive isotopes tritium, i.e. 3 H, and carbon-14, i.e. 14 C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.
  • substitution with heavier isotopes such as deuterium, i.e. 2 H may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances.
  • Substitution with positron emitting isotopes, such as 11 C, 18 F, 15 O and 13 N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.
  • Isotopically-labeled compounds of structure (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Preparations and Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
  • the invention disclosed herein is also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result from, for example, the oxidation, reduction, hydrolysis, amidation, esterification, and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising administering a compound of this invention to a mammal for a period of time sufficient to yield a metabolic product thereof. Such products are typically identified by administering a radiolabeled compound of the invention in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to human, allowing sufficient time for metabolism to occur, and isolating its conversion products from the urine, blood or other biological samples.
  • an animal such as rat, mouse, guinea pig, monkey, or to human
  • Solid compound and “stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • “Mammal” includes humans and both domestic animals such as laboratory animals and household pets (e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like.
  • "Optional” or “optionally” means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not.
  • “optionally substituted aryl” means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
  • “Pharmaceutically acceptable carrier, diluent or excipient” includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
  • “Pharmaceutically acceptable salt” includes both acid and base addition salts.
  • “Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, camphoric acid, camphor- 10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane- 1 ,2-disulfonic acid, ethanesulfonic acid, 2-hydroxyethanesul
  • “Pharmaceutically acceptable base addition salt” refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol,
  • 2-diethylaminoethanol dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, 7V-ethylpiperidine, polyamine resins and the like.
  • Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine. Often crystallizations produce a solvate of the compound of the invention.
  • solvate refers to an aggregate that comprises one or more molecules of a compound of the invention with one or more molecules of solvent.
  • the solvent may be water, in which case the solvate may be a hydrate.
  • the solvent may be an organic solvent.
  • the compounds of the present invention may exist as a hydrate, including a monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding solvated forms.
  • the compound of the invention may be true solvates, while in other cases, the compound of the invention may merely retain adventitious water or be a mixture of water plus some adventitious solvent.
  • a “pharmaceutical composition” refers to a formulation of a compound of the invention and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans.
  • a medium includes all pharmaceutically acceptable carriers, diluents or excipients therefor.
  • Effective amount refers to that amount of a compound of the invention which, when administered to a mammal, preferably a human, is sufficient to effect treatment, as defined below, of a bacterial infection in the mammal, preferably a human.
  • the amount of a compound of the invention which constitutes a “therapeutically effective amount” will vary depending on the compound, the condition and its severity, the manner of administration, and the age of the mammal to be treated, but can be determined routinely by one of ordinary skill in the art having regard to his own knowledge and to this disclosure.
  • Treating covers the treatment of the disease or condition of interest in a mammal, preferably a human, having the disease or condition of interest, and includes:
  • disease or condition i.e., arresting its development
  • relieving the disease or condition i.e., causing regression of the disease or condition
  • relieving the symptoms resulting from the disease or condition i.e., relieving pain without addressing the underlying disease or condition.
  • the terms "disease” and "condition” may be used interchangeably or may be different in that the particular malady or condition may not have a known causative agent (so that etiology has not yet been worked out) and it is therefore not yet recognized as a disease but only as an undesirable condition or syndrome, wherein a more or less specific set of symptoms have been identified by clinicians.
  • the compounds of the invention, or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids.
  • the present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms.
  • Optically active (+) and (-), (R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization.
  • stereoisomer refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable.
  • the present invention contemplates various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another.
  • a “tautomer” refers to a proton shift from one atom of a molecule to another atom of the same molecule.
  • the present invention includes tautomers of any said compounds.
  • compounds having antibacterial activity are provided, the compounds having the following structure (I):
  • Q 1 is hydroxyl, a protected hydroxyl, amino or a protected amino group
  • Q 2 is hydrogen, optionally substituted alkyl
  • Zi and Z 2 are both hydrogen or Zi and Z 2 form a double bond
  • Z 3 and Z 4 are both hydrogen or Z 3 and Z 4 form a double bond.
  • Q 2 is hydrogen
  • each R 4 , R 5 and R 6 independently, hydrogen or Ci-C 6 alkyl, or R 4 and R 5 together with the atoms to which they are attached can form a heterocyclic ring having from 4 to 6 ring atoms, or R 5 and R 6 together with the atoms to which they are attached can form a heterocyclic ring having from 4 to 6 ring atoms, or R 4 and R 6 together with the atoms to which they are attached can form a carbocyclic ring having from 4 to 6 ring atoms.
  • each Ri, R 2 and R 3 are hydrogen.
  • Qi is amino. In other further embodiments, Qi is hydroxyl.
  • Zi and Z 2 form a double bond. In other further embodiments, Zi and Z 2 are both hydrogen.
  • Z 3 and Z 4 form a double bond. In other further embodiments, Z 3 and Z 4 are both hydrogen. In further embodiments, Q 2 is:
  • R 4 is hydrogen; and R 5 is hydrogen.
  • each R 6 is hydrogen.
  • Q 2 is:
  • At least one R 6 is halogen.
  • Q 2 is:
  • R 4 is hydrogen; and R 5 and one R 6 together with the atoms to which they are attached form a heterocyclic ring having from 4 to 6 ring atoms.
  • Q 2 is:
  • At least one R 6 is halogen.
  • Q 2 is: wherein R 4 and R 5 together with the atoms to which they are attached form a heterocyclic ring having from 4 to 6 ring atoms.
  • each R 6 is hydrogen.
  • Q 2 is:
  • At least one R 6 is halogen.
  • Q 2 is:
  • R 5 is hydrogen; and R 4 and one R 6 together with the atoms to which they are attached form a carbocyclic ring having from 3 to 6 ring atoms.
  • Q 2 is:
  • At least one R 6 is halogen.
  • Q 2 is:
  • each R 6 is hydrogen.
  • Q 2 is:
  • At least one R 6 is halogen.
  • Q 2 is:
  • Q 2 is:
  • At least one R 6 is halogen.
  • Q 2 is:
  • R 5 is hydrogen.
  • each R 6 is hydrogen.
  • Q 2 is:
  • At least one R 6 is halogen.
  • Q 2 is:
  • R 7 is hydrogen; and R 8 is hydrogen.
  • each R 6 is hydrogen.
  • Q 2 is:
  • At least one R 6 is halogen.
  • Q 2 is:
  • R 5 is hydrogen.
  • each R 6 is hydrogen.
  • at least one R 6 is halogen.
  • Q 2 is:
  • R 7 is hydrogen; and R 8 is hydrogen.
  • each R 6 is hydrogen.
  • at least one R 6 is halogen.
  • Q 2 is:
  • R 5 is hydrogen.
  • each R 6 is hydrogen.
  • at least one R 6 is halogen.
  • Q 2 is:
  • each Ri 0 is hydrogen. In other further embodiments, at least one Ri 0 is halogen.
  • Q 2 is: wherein: R 7 is hydrogen; and R 8 is hydrogen. In further embodiments, each R] 0 is hydrogen. In other further embodiments, at least one R] 0 is halogen. In other further embodiments, Q 2 is:
  • R 4 is hydrogen.
  • each R 6 is hydrogen.
  • at least one R 6 is halogen.
  • Q 2 is hydrogen. In other further embodiments, Q 2 is optionally substituted alkyl. For example, in more specific embodiments of the foregoing, Q 2 is unsubstituted or Q 2 is substituted with one or more halogen, hydroxyl or amino.
  • any embodiment of the compounds of structure (I), as set forth above, and any specific substituent set forth herein for a Qi, Q 2 , Zi, Z 2 , Z 3 , Z 4 , Ri, R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 and R J 0 group in the compounds of structure (I), as set forth above, may be independently combined with other embodiments and/or substituents of compounds of structure (I) to form embodiments of the inventions not specifically set forth above.
  • compositions of the present invention comprise a compound of structure (I) and a pharmaceutically acceptable carrier, diluent or excipient.
  • the compound of structure (I) is present in the composition in an amount which is effective to treat a particular disease or condition of interest - that is, in an amount sufficient to treat a bacterial infection, and preferably with acceptable toxicity to the patient.
  • the antibacterial activity of compounds of structure (I) can be determined by one skilled in the art, for example, as described in the Examples below. Appropriate concentrations and dosages can be readily determined by one skilled in the art.
  • the compounds of the present invention possess antibacterial activity against a wide spectrum of gram positive and gram negative bacteria, as well as enterobacteria and anaerobes.
  • Representative susceptible organisms generally include those gram positive and gram negative, aerobic and anaerobic organisms whose growth can be inhibited by the compounds of the invention such as Staphylococcus, Lactobacillus, Streptococcus, Sarcina, Escherichia, Enterobacter, Klebsiella, Pseudomonas, Acinetobacter, Mycobacterium, Proteus, Campylobacter, Citrobacter, Nisseria, Baccillus, Bacteroides, Peptococcus, Clostridium, Salmonella, Shigella, Serratia, Haemophilus, Brucella and other organisms.
  • compositions of the invention can be prepared by combining a compound of the invention with an appropriate pharmaceutically acceptable carrier, diluent or excipient, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols.
  • compositions of the invention are formulated so as to allow the active ingredients contained therein to be bioavailable upon administration of the composition to a patient.
  • Compositions that will be administered to a subject or patient take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of a compound of the invention in aerosol form may hold a plurality of dosage units.
  • compositions to be administered will, in any event, contain a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, for treatment of a disease or condition of interest in accordance with the teachings of this invention.
  • a pharmaceutical composition of the invention may be in the form of a solid or liquid.
  • the carrier(s) are particulate, so that the compositions are, for example, in tablet or powder form.
  • the carrier(s) may be liquid, with the compositions being, for example, an oral syrup, injectable liquid or an aerosol, which is useful in, for example, inhalatory administration.
  • the pharmaceutical composition is preferably in either solid or liquid form, where semi-solid, semi-liquid, suspension and gel forms are included within the forms considered herein as either solid or liquid.
  • the pharmaceutical composition may be formulated into a powder, granule, compressed tablet, pill, capsule, chewing gum, wafer or the like form.
  • a solid composition will typically contain one or more inert diluents or edible carriers.
  • binders such as carboxymethylcellulose, ethyl cellulose, microcrystalline cellulose, gum tragacanth or gelatin; excipients such as starch, lactose or dextrins, disintegrating agents such as alginic acid, sodium alginate, Primogel, corn starch and the like; lubricants such as magnesium stearate or Sterotex; glidants such as colloidal silicon dioxide; sweetening agents such as sucrose or saccharin; a flavoring agent such as peppermint, methyl salicylate or orange flavoring; and a coloring agent.
  • excipients such as starch, lactose or dextrins, disintegrating agents such as alginic acid, sodium alginate, Primogel, corn starch and the like
  • lubricants such as magnesium stearate or Sterotex
  • glidants such as colloidal silicon dioxide
  • sweetening agents such as sucrose or saccharin
  • a flavoring agent such as peppermint, methyl sal
  • the pharmaceutical composition when in the form of a capsule, for example, a gelatin capsule, it may contain, in addition to materials of the above type, a liquid carrier such as polyethylene glycol or oil.
  • a liquid carrier such as polyethylene glycol or oil.
  • the pharmaceutical composition may be in the form of a liquid, for example, an elixir, syrup, solution, emulsion or suspension.
  • the liquid may be for oral administration or for delivery by injection, as two examples.
  • preferred composition contain, in addition to the present compounds, one or more of a sweetening agent, preservatives, dye/colorant and flavor enhancer.
  • a surfactant, preservative, wetting agent, dispersing agent, suspending agent, buffer, stabilizer and isotonic agent may be included.
  • the liquid pharmaceutical compositions of the invention may include one or more of the following adjuvants: sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils such as synthetic mono or diglycerides which may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride
  • fixed oils such as synthetic mono or diglycerides which may
  • parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • Physiological saline is a preferred adjuvant.
  • An injectable pharmaceutical composition is preferably sterile.
  • a liquid pharmaceutical composition of the invention intended for either parenteral or oral administration should contain an amount of a compound of the invention such that a suitable dosage will be obtained.
  • the pharmaceutical composition of the invention may be intended for topical administration, in which case the carrier may suitably comprise a solution, emulsion, ointment or gel base.
  • the base for example, may comprise one or more of the following: petrolatum, lanolin, polyethylene glycols, bee wax, mineral oil, diluents such as water and alcohol, and emulsif ⁇ ers and stabilizers.
  • Thickening agents may be present in a pharmaceutical composition for topical administration.
  • the composition may include a transdermal patch or iontophoresis device.
  • the pharmaceutical composition of the invention may be intended for rectal administration, in the form, for example, of a suppository, which will melt in the rectum and release the drug.
  • the composition for rectal administration may contain an oleaginous base as a suitable nonirritating excipient.
  • bases include, without limitation, lanolin, cocoa butter and polyethylene glycol.
  • the pharmaceutical composition of the invention may include various materials, which modify the physical form of a solid or liquid dosage unit.
  • the composition may include materials that form a coating shell around the active ingredients.
  • the materials that form the coating shell are typically inert, and may be selected from, for example, sugar, shellac, and other enteric coating agents.
  • the active ingredients may be encased in a gelatin capsule.
  • the pharmaceutical composition of the invention in solid or liquid form may include an agent that binds to the compound of the invention and thereby assists in the delivery of the compound.
  • Suitable agents that may act in this capacity include a monoclonal or polyclonal antibody, a protein or a liposome.
  • the pharmaceutical composition of the invention may consist of dosage units that can be administered as an aerosol.
  • aerosol is used to denote a variety of systems ranging from those of colloidal nature to systems consisting of pressurized packages. Delivery may be by a liquefied or compressed gas or by a suitable pump system that dispenses the active ingredients. Aerosols of compounds of the invention may be delivered in single phase, bi-phasic, or tri-phasic systems in order to deliver the active ingredient(s). Delivery of the aerosol includes the necessary container, activators, valves, subcontainers, and the like, which together may form a kit. One skilled in the art, without undue experimentation may determine preferred aerosols.
  • the pharmaceutical compositions of the invention may be prepared by methodology well known in the pharmaceutical art.
  • a pharmaceutical composition intended to be administered by injection can be prepared by combining a compound of the invention with sterile, distilled water so as to form a solution.
  • a surfactant may be added to facilitate the formation of a homogeneous solution or suspension.
  • Surfactants are compounds that non-covalently interact with the compound of the invention so as to facilitate dissolution or homogeneous suspension of the compound in the aqueous delivery system.
  • the compounds of the invention are administered in a therapeutically effective amount, which will vary depending upon a variety of factors including the activity of the specific compound employed; the metabolic stability and length of action of the compound; the age, body weight, general health, sex, and diet of the patient; the mode and time of administration; the rate of excretion; the drug combination; the severity of the particular disorder or condition; and the subject undergoing therapy.
  • Compounds of the invention, or pharmaceutically acceptable derivatives thereof, may also be administered simultaneously with, prior to, or after administration of one or more other therapeutic agents.
  • Such combination therapy includes administration of a single pharmaceutical dosage formulation which contains a compound of the invention and one or more additional active agents, as well as administration of the compound of the invention and each active agent in its own separate pharmaceutical dosage formulation.
  • a compound of the invention and the other active agent can be administered to the patient together in a single oral dosage composition such as a tablet or capsule, or each agent administered in separate oral dosage formulations.
  • the compounds of the invention and one or more additional active agents can be administered at essentially the same time, i.e., concurrently, or at separately staggered times, i.e., sequentially; combination therapy is understood to include all these regimens.
  • suitable protecting groups include hydroxy, amino, mercapto and carboxylic acid.
  • suitable protecting groups for hydroxy include trialkylsilyl or diarylalkylsilyl (for example, f-butyldimethylsilyl, t- butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like
  • suitable protecting groups for amino, amidino and guanidino include r-butoxycarbonyl, benzyloxycarbonyl, and the like.
  • Suitable protecting groups for mercapto include -C(O)-R" (where R" is alkyl, aryl or arylalkyl), /?-methoxybenzyl, trityl and the like.
  • Suitable protecting groups for carboxylic acid include alkyl, aryl or arylalkyl esters.
  • Protecting groups may be added or removed in accordance with standard techniques, which are known to one skilled in the art and as described herein. The use of protecting groups is described in detail in Green, T.W. and P. G. M. Wutz, Protective Groups in Organic Synthesis (1999), 3rd Ed., Wiley.
  • the protecting group may also be a polymer resin such as a Wang resin, Rink resin or a 2-chlorotrityl-chloride resin.
  • Paromomycin sulfate (6, 685 g free base, 1113 mmol) was dissolved in water (4 L) and then ACN (4 L) was slowly added to the reaction. The reaction was cooled to O 0 C and then DIPEA (1525 mL, 8910 mmol) was added. O-Cbz-2- nitrophenol (5, 304 g, 1113 mmol) was dissolved in ACN (1 L) and added to the reaction via an addition funnel in one hour.
  • reaction mixture was then diluted with ethyl acetate, washed with water, dried over Na 2 SO 4 and concentrated to a yellow oil, which was purified on a 1- inch reverse-phase HPLC column to yield 3',4',3'",4"'-tetradehydro-6'-benzyl-l-(7V- Cbz-4-amino-2(,S)-hydroxy-butyryl)-per-Cbz-paromomycin (15): MS: m/z (M+Na) + calc. 1431.56, obs. 1431.5.
  • Neomycin sulfate (1, 240 g, 391 mmol, 1 eq) was suspended in MeOH
  • reaction vessel 250 mg of Pd(OH) 2 on carbon (20% dry basis Pd), and the reaction vessel was evacuated and replenished with hydrogen. The reaction was monitored by HPLC-MS and deemed complete after 1 hour. The reaction mixture was filtered through a .45 ⁇ m
  • aqueous layer was washed with 200 mL ethyl acetate and the combined organics were then washed with 600 mL of sat. aq. NaHCO 3 followed by 600 mL of a 1 :1 mixture of water and brine.
  • reaction solution was filtered through Celite that had been rinsed with 200 mL water and the cake was rinsed with an additional 500 mL water.
  • the filtrate was diluted to a final volume of 1.8 L with water and then lyophilized.
  • the crude solid was purified on a 2-inch reverse-phase HPLC column (0-10% ACN, 10 niM NH 4 OH) to yield 3',4',3'",4'"-tetra-dehydro-l-(4-amino-2(5)-hydroxy-butyryl)-neomycin (5, 1.87 g, 74.8% yield): MS: m/z (M+Na) + calc.674.38; obs. 674.4.
  • the aqueous layer was acidified with acetic acid (0.5 mL) and washed with ethyl acetate (2 x 30 mL). The aqueous layer was frozen and lyophilized, then purified by preparative HPLC. The appropriate fractions were combined and the desired product was free-based with IRA-400 (OH form) ion-exchange resin, then filtered and lyophilized to yield (7) as a white solid (6 mg, 0.009 mmol, 50%). Calculated for C 27 H 49 N 7 O n : 670.35 (M+Na) + ; found: 670.3.
  • Neomycin sulfate (1, 240 g, 391 mmol, 1 eq, mixture of B/C isomers) was suspended in methanol (10 L) then was diluted with water (400 mL). The reaction was cooled to O 0 C and K 2 CO 3 (1080 g, 7818 mmol, 20 eq) was added slowly to the reaction mixture. Next benzyl chloro formate (550 mL, 3910 mmol, 10 eq) was added to the reaction mixture via a dropping funnel over 3 hours. The mixture was stirred vigorously for 18 hours at room temperature, then was filtered and concentrated to 2 L.
  • Procedure 1 Hydro genolysis To a stirring solution of aminoglycoside (0.41 mmol) in AcOH (60 mL) was added H 2 O (15 mL), followed by Pd(OH) 2 /C (30% by weight, 50% H 2 O). The reaction vessel was evacuated and replenished with H 2 (1 atm), and the reaction mixture was stirred for 4 hr. The solids were removed by filtration through a pad of Celite, and washed with H 2 O (100 mL). The aqueous layer was lyophilized to yield the desired product.
  • reaction mixture was then purified on a 2-inch reverse-phase HPLC column to yield 3',4',3'",4'"-tetra-dehydro-l-(N-Boc-2-hydroxy-3-amino-propyl)-3,2',6',2'",6'"-penta- Cbz-neomycin (2, 115 mg, 0.083 mmol, 49.4% yield): MS: m/z (M+Na) + calc. 1412.59, obs.: 1412.4.
  • reaction mixture was then purified on a 2-inch reverse-phase HPLC column to yield 3',4',3'",4'"-tetra-dehydro-l- (3-cyano-propionyl)-3,2',6',2'",6'"-penta-Cbz-neomycin (105 mg, 51% yield): MS: m/z (M+Na) + calc. 1320.5, obs. 1320.4.
  • reaction mixture was purified on a 2-inch reverse- phase HPLC column to yield 3',4',3'",4'"-tetra-dehydro-l-(3-hydroxybenzyl)- 3,2 ⁇ 6 ⁇ 2"',6"'-penta-Cbz-neomycin (2, 140 mg, 0.106 mmol, 66% yield): MS: m/z (M+H) + calc. 1323.5, obs. 1323.3.
  • reaction mixture was purified on a 2-inch reverse-phase HPLC column to yield 3',4',3'",4'"-tetra-dehydro-l-(2,3-bis-hydroxy-propyl)-3,2',6',2'",6'"-penta-Cbz- neomycin (2, 26 mg, 0.020 mmol, 13 % yield): MS: m/z (M+H) + calc. 1291.5, obs.1291.3.
  • reaction mixture was purified on a 2-inch reverse-phase HPLC column to yield 3',4',3'",4'"-tetra-dehydro-l-(N-Cbz-2-amino-acetyl)-3,2',6',2'",6'"-penta-Cbz- neomycin (2, 220 mg, 0.156 mmol, 95 % yield): MS: m/z (M+Na) + calc. 1430.54, obs. 1430.4.
  • reaction mixture was purified on a 2-inch reverse-phase HPLC column to yield 3',4',3'",4'"-tetra-dehydro-l-(iV-Cbz-2( ( S)-ammo-3-hydroxy-propionyl)- 3,2',6',2'",6'"- penta-Cbz-neomycin (2, 175 mg, 0.122 mmol, 74.4 % yield): MS: m/z (M+Na) + calc. 1460.55, obs.1460.5.
  • the reaction was then diluted with EtOAc (10 mL), and the aqueous phase was separated. The organic layer was washed with sat. aq. NaHCO 3 (10 mL). The aqueous layers were back-extracted with EtOAc (10 mL). The combined organic layers were washed with sat. aq. NaHCO 3 (10 mL), sat. aq.
  • Penta-l,4-dienol (5 g, 59.4 mmol) and excess cumene hydroperoxide (80%, 17.5 mL) were added in small portions, and stirring was continued at -35°C for 48 hr.
  • the reaction was quenched by addition of sat. aq. Na 2 SO 4 (5 mL) immediately followed by Et 2 O (50 mL) and the reaction was stirred for 2 hr with warming to rt.
  • the reaction mixture was filtered through Celite, and washed with Et 2 O. Solvent removal under vacuum without heating resulted in approximately 30 mL of a yellow solution.
  • iV-Methyl morpholine (1.41 mL, 12.9 mmol) was added dropwise, and the reaction was stirred at -15°C for 2 hr. The reaction was quenched with phosphate buffer (0.1 M, pH 6.0) and the aqueous layer was separated. The organic layer was washed with the phosphate buffer (3 x), dried over Na 2 SO 4 , filtered and reduced under vacuum to give a brown residue.
  • phosphate buffer 0.1 M, pH 6.0
  • MIC Minimum inhibitory concentrations
  • CLSI Clinical and Laboratory Standards Institute
  • Quality control ranges utilizing E. coli ATCC 25922, P. aeruginosa ATCC 27853 and S. aureus ATCC 29213, and interpretive criteria for comparator agents were as published in CLSI Ml 00-Sl 7 [2007].
  • serial two-fold dilutions of the test compounds were prepared at 2X concentration in Mueller Hinton Broth.
  • the compound dilutions were mixed in 96-well assay plates in a 1 :1 ratio with bacterial inoculum.
  • the inoculum was prepared by suspension of a colony from an agar plate that was prepared the previous day.

Abstract

Compounds having antibacterial activity are disclosed. The compounds have the following structure (I) including stereoisomers, pharmaceutically acceptable salts and prodrugs thereof, wherein Q1, Q2, Z1, Z2, Z3, Z4, R1, R2 and R3 are as defined herein. Methods associated with preparation and use of such compounds, as well as pharmaceutical compositions comprising such compounds, are also disclosed.

Description

ANTIBACTERIAL AMINOGLYCOSIDE ANALOGS
CROSS-REFERENCE TO RELATED APPLICATION
This application claims the benefit under 35 U. S. C. §119(e) of U.S. Provisional Patent Application No. 61/095,673, filed September 10, 2008, which application is incorporated herein by reference in its entirety.
BACKGROUND OF THE INVENTION
Field of the Invention
The present invention is directed to novel aminoglycoside compounds, and methods for their preparation and use as therapeutic or prophylactic agents.
Description of the Related Art A particular interest in modern drug discovery is the development of novel low molecular weight orally-bioavailable drugs that work by binding to RNA. RNA, which serves as a messenger between DNA and proteins, was thought to be an entirely flexible molecule without significant structural complexity. Recent studies have revealed a surprising intricacy in RNA structure. RNA has a structural complexity rivaling proteins, rather than simple motifs like DNA. Genome sequencing reveals both the sequences of the proteins and the mRNAs that encode them. Since proteins are synthesized using an RNA template, such proteins can be inhibited by preventing their production in the first place by interfering with the translation of the mRNA. Since both proteins and the RNAs are potential drug targeting sites, the number of targets revealed from genome sequencing efforts is effectively doubled. These observations unlock a new world of opportunities for the pharmaceutical industry to target RNA with small molecules.
Classical drug discovery has focused on proteins as targets for intervention. Proteins can be extremely difficult to isolate and purify in the appropriate form for use in assays for drug screening. Many proteins require post-translational modifications that occur only in specific cell types under specific conditions. Proteins fold into globular domains with hydrophobic cores and hydrophilic and charged groups on the surface. Multiple subunits frequently form complexes, which may be required for a valid drug screen. Membrane proteins usually need to be embedded in a membrane to retain their proper shape. The smallest practical unit of a protein that can be used in drug screening is a globular domain. The notion of removing a single alpha helix or turn of a beta sheet and using it in a drug screen is not practical, since only the intact protein may have the appropriate 3 -dimensional shape for drug binding. Preparation of biologically active proteins for screening is a major limitation in classical high throughput screening. Quite often the limiting reagent in high throughput screening efforts is a biologically active form of a protein which can also be quite expensive.
For screening to discover compounds that bind RNA targets, the classic approaches used for proteins can be superceded with new approaches. All RNAs are essentially equivalent in their solubility, ease of synthesis or use in assays. The physical properties of RNAs are independent of the protein they encode. They may be readily prepared in large quantity through either chemical or enzymatic synthesis and are not extensively modified in vivo. With RNA, the smallest practical unit for drug binding is the functional subdomain. A functional subdomain in RNA is a fragment that, when removed from the larger RNA and studied in isolation, retains its biologically relevant shape and protein or RNA-binding properties. The size and composition of RNA functional subdomains make them accessible by enzymatic or chemical synthesis. The structural biology community has developed significant experience in identification of functional RNA subdomains in order to facilitate structural studies by techniques such as NMR spectroscopy. For example, small analogs of the decoding region of 16S rRNA (the A-site) have been identified as containing only the essential region, and have been shown to bind antibiotics in the same fashion as the intact ribosome.
The binding sites on RNA are hydrophilic and relatively open as compared to proteins. The potential for small molecule recognition based on shape is enhanced by the deformability of RNA. The binding of molecules to specific RNA targets can be determined by global conformation and the distribution of charged, aromatic, and hydrogen bonding groups off of a relatively rigid scaffold. Properly placed positive charges are believed to be important, since long-range electrostatic interactions can be used to steer molecules into a binding pocket with the proper orientation. In structures where nucleobases are exposed, stacking interactions with aromatic functional groups may contribute to the binding interaction. The major groove of RNA provides many sites for specific hydrogen bonding with a ligand. These include the aromatic N7 nitrogen atoms of adenosine and guanosine, the 04 and O6 oxygen atoms of undine and guanosine, and the amines of adenosine and cytidine. The rich structural and sequence diversity of RNA suggests to us that ligands can be created with high affinity and specificity for their target.
Although our understanding of RNA structure and folding, as well as the modes in which RNA is recognized by other ligands, is far from being comprehensive, significant progress has been made in the last decade (see, e.g., Chow, C. S.; Bogdan, F.M., Chem. Rev., 1997, 97, 1489 and Wallis, M.G.; Schroeder, R., Prog. Biophys. Molec. Biol. 1997, 67, 141). Despite the central role RNA plays in the replication of bacteria, drugs that target these pivotal RNA sites of these pathogens are scarce. The increasing problem of bacterial resistance to antibiotics makes the search for novel RNA binders of crucial importance.
Certain small molecules can bind and block essential functions of RNA. Examples of such molecules include the aminoglycoside antibiotics and drugs such as erythromycin which binds to bacterial rRNA and releases peptidyl-tRNA and mRNA. Aminoglycoside antibiotics have long been known to bind RNA. They exert their antibacterial effects by binding to specific target sites in the bacterial ribosome. For the structurally related antibiotics neamine, ribostamycin, neomycin B, and paromomycin, the binding site has been localized to the A-site of the prokaryotic 16S ribosomal decoding region RNA (see Moazed, D.; Noller, H.F., Nature, 1987, 327, 389). Binding of aminoglycosides to this RNA target interferes with the fidelity of mRNA translation and results in miscoding and truncation, leading ultimately to bacterial cell death (see Alper, P.B.; Hendrix, M.; Sears, P.; Wong, C, J Am. Chem. Soc, 1998, 120, 1965). There is a need in the art for new chemical entities that work against bacteria with broad-spectrum activity. Perhaps the biggest challenge in discovering RNA-binding antibacterial drugs is identifying vital structures common to bacteria that can be disabled by small molecule drug binding. A challenge in targeting RNA with small molecules is to develop a chemical strategy which recognizes specific shapes of RNA. There are three sets of data that provide hints on how to do this: natural protein interactions with RNA, natural product antibiotics that bind RNA, and man-made RNAs (aptamers) that bind proteins and other molecules. Each data set, however, provides different insights to the problem.
Several classes of drugs obtained from natural sources have been shown to work by binding to RNA or RNA/protein complexes. These include three different structural classes of antibiotics: thiostreptone, the aminoglycoside family and the macrolide family of antibiotics. These examples provide powerful clues to how small molecules and targets might be selected. Nature has selected RNA targets in the ribosome, one of the most ancient and conserved targets in bacteria. Since antibacterial drugs are desired to be potent and have broad-spectrum activity, these ancient processes, fundamental to all bacterial life, represent attractive targets. The closer we get to ancient conserved functions the more likely we are to find broadly conserved RNA shapes. It is important to also consider the shape of the equivalent structure in humans, since bacteria were unlikely to have considered the therapeutic index of their RNAs while evolving them. A large number of natural antibiotics exist, these include the aminoglycosides, such as, kirromycin, neomycin, paromomycin, thiostrepton, and many others. They are very potent, bactericidal compounds that bind RNA of the small ribosomal subunit. The bactericidal action is mediated by binding to the bacterial RNA in a fashion that leads to misreading of the genetic code. Misreading of the code during translation of integral membrane proteins is thought to produce abnormal proteins that compromise the barrier properties of the bacterial membrane.
Antibiotics are chemical substances produced by various species of microorganisms (bacteria, fungi, actinomycetes) that suppress the growth of other microorganisms and may eventually destroy them. However, common usage often extends the term antibiotics to include synthetic antibacterial agents, such as the sulfonamides, and quinolines, that are not products of microbes. The number of antibiotics that have been identified now extends into the hundreds, and many of these have been developed to the stage where they are of value in the therapy of infectious diseases. Antibiotics differ markedly in physical, chemical, and pharmacological properties, antibacterial spectra, and mechanisms of action. In recent years, knowledge of molecular mechanisms of bacterial, fungal, and viral replication has greatly facilitated rational development of compounds that can interfere with the life cycles of these microorganisms.
At least 30% of all hospitalized patients now receive one or more courses of therapy with antibiotics, and millions of potentially fatal infections have been cured. At the same time, these pharmaceutical agents have become among the most misused of those available to the practicing physician. One result of widespread use of antimicrobial agents has been the emergence of antibiotic-resistant pathogens, which in turn has created an ever-increasing need for new drugs. Many of these agents have also contributed significantly to the rising costs of medical care. When the antimicrobial activity of a new agent is first tested, a pattern of sensitivity and resistance is usually defined. Unfortunately, this spectrum of activity can subsequently change to a remarkable degree, because microorganisms have evolved the array of ingenious alterations discussed above that allow them to survive in the presence of antibiotics. The mechanism of drug resistance varies from microorganism to microorganism and from drug to drug.
The development of resistance to antibiotics usually involves a stable genetic change, inheritable from generation to generation. Any of the mechanisms that result in alteration of bacterial genetic composition can operate. While mutation is frequently the cause, resistance to antimicrobial agents may be acquired through transfer of genetic material from one bacterium to another by transduction, transformation or conjugation.
For the foregoing reasons, while progress has been made in this field, there is a need for new chemical entities that possess antibacterial activity. Further, in order to accelerate the drug discovery process, new methods for synthesizing aminoglycoside antibiotics are needed to provide an array of compounds that are potentially new drugs for the treatment of bacterial infections. The present invention fulfills these needs and provides further related advantages.
BRIEF SUMMARY OF THE INVENTION In brief, the present invention is directed to novel aminoglycoside compounds, having antibacterial activity, including stereoisomers, pharmaceutically acceptable salts and prodrugs thereof, and the use of such compounds in the treatment of bacterial infections.
In one embodiment, compounds having the following structure (I) are provided:
Figure imgf000008_0001
(I) or a stereoisomer, pharmaceutically acceptable salt or prodrug thereof, wherein: wherein: Qi is hydroxyl, a protected hydroxyl, amino or a protected amino group;
Q2 is hydrogen, optionally substituted alkyl,
Figure imgf000008_0002
Figure imgf000009_0001
Figure imgf000009_0002
Figure imgf000009_0003
Figure imgf000009_0004
10
Figure imgf000009_0005
Figure imgf000010_0001
Figure imgf000010_0002
each Ri and R2 is, independently, hydrogen or an amino protecting group; each R3 is, independently, hydrogen or a hydroxyl protecting group; each R4, R5, R7 and R8 is, independently, hydrogen or Cj-C6 alkyl optionally substituted with one or more halogen, hydroxyl or amino; each R6 is, independently, hydrogen, halogen, hydroxyl, amino or C]-C6 alkyl; or R4 and R5 together with the atoms to which they are attached can form a heterocyclic ring having from 4 to 6 ring atoms, or R5 and one R6 together with the atoms to which they are attached can form a heterocyclic ring having from 4 to 6 ring atoms, or R4 and one R6 together with the atoms to which they are attached can form a carbocyclic ring having from 3 to 6 ring atoms, or R7 and R8 together with the atom to which they are attached can form a heterocyclic ring having from 4 to 6 ring atoms; each R9 is, independently, hydrogen, hydroxyl, amino or Ci-C6 alkyl; each Rio is, independently, hydrogen, halogen, hydroxyl, amino or CpC6 alkyl; or R.9 and one Rio together with the atoms to which they are attached can form a heterocyclic ring having from 4 to 6 ring atoms; n is an integer from 0 to 4;
Zi and Z2 are both hydrogen or Zj and Z2 form a double bond; and
Z3 and Z4 are both hydrogen or Z3 and Z4 form a double bond.
In another embodiment, a pharmaceutical composition is provided comprising a compound having structure (I), or a stereoisomer, pharmaceutically acceptable salt or prodrug thereof, and a pharmaceutically acceptable carrier, diluent or excipient.
In another embodiment, a method of using a compound having structure
(I) in therapy is provided. In particular, the present invention provides a method of treating a bacterial infection in a mammal comprising administering to the mammal an effective amount of a compound having structure (I), or a stereoisomer, pharmaceutically acceptable salt or prodrug thereof.
These and other aspects of the invention will be apparent upon reference to the following detailed description.
DETAILED DESCRIPTION OF THE INVENTION
In the following description, certain specific details are set forth in order to provide a thorough understanding of various embodiments of the invention.
However, one skilled in the art will understand that the invention may be practiced without these details. Unless the context requires otherwise, throughout the present specification and claims, the word "comprise" and variations thereof, such as,
"comprises" and "comprising" are to be construed in an open, inclusive sense, that is as
"including, but not limited to".
Reference throughout this specification to "one embodiment" or "an embodiment" means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment of the present invention. Thus, the appearances of the phrases "in one embodiment" or "in an embodiment" in various places throughout this specification are not necessarily all referring to the same embodiment. Furthermore, the particular features, structures, or characteristics may be combined in any suitable manner in one or more embodiments. "Amino" refers to the -NH2 radical.
"Cyano" refers to the -CN radical.
"Hydroxy" or "hydroxyl" refers to the -OH radical.
"Imino" refers to the =NH substituent.
"Nitro" refers to the -NO2 radical. "Oxo" refers to the =0 substituent.
"Thioxo" refers to the =S substituent.
"Alkyl" refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds), having from one to twelve carbon atoms (Ci-Ci2 alkyl), preferably one to eight carbon atoms (C)-Cg alkyl) or one to six carbon atoms (Ci-C6 alkyl), and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, n-propyl, 1 -methyl ethyl (zsø-propyl), «-butyl, n-pentyl,
1,1 -dimethyl ethyl (f-butyl), 3-methylhexyl, 2-methylhexyl, ethenyl, prop-1-enyl, but-1-enyl, pent-1-enyl, penta-l,4-dienyl, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the specification, an alkyl group may be optionally substituted.
"Alkylene" or "alkylene chain" refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds), and having from one to twelve carbon atoms, e.g., methylene, ethylene, propylene, rc-butylene, ethenylene, propenylene, w-butenylene, propynylene, n-butynylene, and the like. The alkylene chain is attached to the rest of the molecule through a single or double bond and to the radical group through a single or double bond. The points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, an alkylene chain may be optionally substituted.
"Alkoxy" refers to a radical of the formula -ORa where R3 is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkoxy group may be optionally substituted.
"Alkylamino" refers to a radical of the formula -NHRa or -NR3R3 where each R3 is, independently, an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkylamino group may be optionally substituted.
"Thioalkyl" refers to a radical of the formula -SR3 where R3 is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, a thioalkyl group may be optionally substituted. "Aryl" refers to a hydrocarbon ring system radical comprising hydrogen,
6 to 18 carbon atoms and at least one aromatic ring. For purposes of this invention, the aryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems. Aryl radicals include, but are not limited to, aryl radicals derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, <zs-indacene, s-indacene, indane, indene, naphthalene, phenalene, phenanthrene, pleiadene, pyrene, and triphenylene. Unless stated otherwise specifically in the specification, the term "aryl" or the prefix "ar-" (such as in "aralkyl") is meant to include aryl radicals that are optionally substituted. "Aralkyl" refers to a radical of the formula -Rb-Rc where Rb is an alkylene chain as defined above and Rc is one or more aryl radicals as defined above, for example, benzyl, diphenylmethyl and the like. Unless stated otherwise specifically in the specification, an aralkyl group may be optionally substituted.
"Cycloalkyl" or "carbocyclic ring" refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which may include fused or bridged ring systems, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond. Monocyclic radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Polycyclic radicals include, for example, adamantyl, norbornyl, decalinyl, 7,7-dimethyl-bicyclo[2.2.1]heptanyl, and the like. Unless otherwise stated specifically in the specification, a cycloalkyl group may be optionally substituted.
"Cycloalkylalkyl" refers to a radical of the formula -RbRa where Ra is an alkylene chain as defined above and Rg is a cycloalkyl radical as defined above. Unless stated otherwise specifically in the specification, a cycloalkylalkyl group may be optionally substituted.
"Fused" refers to any ring structure described herein which is fused to an existing ring structure in the compounds of the invention. When the fused ring is a heterocyclyl ring or a heteroaryl ring, any carbon atom on the existing ring structure which becomes part of the fused heterocyclyl ring or the fused heteroaryl ring may be replaced with a nitrogen atom.
"Halo" or "halogen" refers to bromo, chloro, fluoro or iodo. "Haloalkyl" refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1,2-difluoroethyl,
3-bromo-2-fluoropropyl, 1 ,2-dibromoethyl, and the like. Unless stated otherwise specifically in the specification, a haloalkyl group may be optionally substituted.
"Heterocyclyl" or "heterocyclic ring" refers to a stable 3- to 18-membered non-aromatic ring radical which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. Unless stated otherwise specifically in the specification, the heterocyclyl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized; and the heterocyclyl radical may be partially or fully saturated. Examples of such heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl[l,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, moφholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1 -oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl. Unless stated otherwise specifically in the specification, a heterocyclyl group may be optionally substituted.
'W-heterocyclyl" refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a nitrogen atom in the heterocyclyl radical. Unless stated otherwise specifically in the specification, a 7V-heterocyclyl group may be optionally substituted.
"Heterocyclylalkyl" refers to a radical of the formula -RbRe where Rb is an alkylene chain as defined above and Re is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl may be attached to the alkyl radical at the nitrogen atom. Unless stated otherwise specifically in the specification, a heterocyclylalkyl group may be optionally substituted.
"Heteroaryl" refers to a 5- to 14-membered ring system radical comprising hydrogen atoms, one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and at least one aromatic ring. For purposes of this invention, the heteroaryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized. Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzothiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[ό][l,4]dioxepinyl, 1 ,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[l,2-a]pyridinyl, carbazolyl, cinnolinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furanonyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1- oxidopyridinyl, 1-oxidopyrimidinyl, 1-oxidopyrazinyl, 1-oxidopyridazinyl, 1-phenyl-lH-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinazolinyl, quinoxalinyl, quinolinyl, quinuclidinyl, isoquinolinyl, tetrahydroquinolinyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, triazinyl, and thiophenyl (i.e. thienyl). Unless stated otherwise specifically in the specification, a heteroaryl group may be optionally substituted. 'W-heteroaryl" refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical. Unless stated otherwise specifically in the specification, an 7V-heteroaryl group may be optionally substituted. "Ηeteroarylalkyl" refers to a radical of the formula -RbRf where Rb is an alkylene chain as defined above and Rf is a heteroaryl radical as defined above. Unless stated otherwise specifically in the specification, a heteroarylalkyl group may be optionally substituted.
The term "substituted" used herein means any of the above groups (i.e., alkyl, alkylene, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, 7V-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl) wherein at least one hydrogen atom is replaced by a bond to a non-hydrogen atoms such as, but not limited to: a halogen atom such as F, Cl, Br, and I; an oxygen atom in groups such as hydroxyl groups, alkoxy groups, and ester groups; a sulfur atom in groups such as thiol groups, thioalkyl groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, imides, and enamines; a silicon atom in groups such as trialkylsilyl groups, dialkylarylsilyl groups, alkyldiarylsilyl groups, and triarylsilyl groups; and other heteroatoms in various other groups. "Substituted" also means any of the above groups in which one or more hydrogen atoms are replaced by a higher-order bond (e.g., a double- or triple-bond) to a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and ester groups; and nitrogen in groups such as imines, oximes, hydrazones, and nitriles. For example, "substituted" includes any of the above groups in which one or more hydrogen atoms are replaced with -NRgRh, -NRgC(=O)Rh, -NRgC(=O)NRgRh, -NRgC(=O)ORh, -NRgSO2Rh, -OC(=O)NRgRh, -ORg, -SRg, -SORg, -SO2Rg, -OSO2Rg, -SO2ORg, =NSO2Rg, and -SO2NRgRi1. "Substituted also means any of the above groups in which one or more hydrogen atoms are replaced with -C(=O)Rg, -C(=O)ORg, -C(=O)NRgRh, -CH2SO2Rg, -CH2SO2NRgRh. In the foregoing, Rg and Rh are the same or different and independently hydrogen, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, JV-heterocyclyl, heterocyclylalkyl, heteroaryl, 7V-heteroaryl and/or heteroarylalkyl. "Substituted" further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to an amino, cyano, hydroxyl, imino, nitro, oxo, thioxo, halo, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, 7V-heterocyclyl, heterocyclylalkyl, heteroaryl, 7V-heteroaryl and/or heteroarylalkyl group. In addition, each of the foregoing substituents may also be optionally substituted with one or more of the above substituents.
The term "protecting group," as used herein, refers to a labile chemical moiety which is known in the art to protect reactive groups including without limitation, hydroxyl and amino groups, against undesired reactions during synthetic procedures. Hydroxyl and amino groups which protected with a protecting group are referred to herein as "protected hydroxyl groups" and "protected amino groups", respectively. Protecting groups are typically used selectively and/or orthogonally to protect sites during reactions at other reactive sites and can then be removed to leave the unprotected group as is or available for further reactions. Protecting groups as known in the art are described generally in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, New York (1999). Groups can be selectively incorporated into aminoglycosides of the invention as precursors. For example an amino group can be placed into a compound of the invention as an azido group that can be chemically converted to the amino group at a desired point in the synthesis. Generally, groups are protected or present as a precursor that will be inert to reactions that modify other areas of the parent molecule for conversion into their final groups at an appropriate time. Further representative protecting or precursor groups are discussed in Agrawal, et al., Protocols for Oligonucleotide Conjugates, Eds, Humana Press; New Jersey, 1994; Vol. 26 pp. 1 -72. Examples of "hydroxyl protecting groups" include, but are not limited to, t-butyl, t-butoxymethyl, methoxymethyl, tetrahydropyranyl, 1-ethoxyethyl, l-(2- chloroethoxy)ethyl, 2-trimethylsilylethyl, p-chlorophenyl, 2,4-dinitrophenyl, benzyl, 2,6-dichlorobenzyl, diphenylmethyl, p-nitrobenzyl, triphenylmethyl, trimethylsilyl, triethylsilyl, t-butyldimethylsilyl, t-butyldiphenylsilyl, triphenylsilyl, benzoylformate, acetate, chloroacetate, trichloroacetate, trifluoroacetate, pivaloate, benzoate, p- phenylbenzoate, 9-fluorenylmethyl carbonate, mesylate and tosylate. Examples of "amino protecting groups" include, but are not limited to, carbamate-protecting groups, such as 2-trimethylsilylethoxycarbonyl (Teoc), 1 -methyl- 1 -(4-biphenyl yl)ethoxy- carbonyl (Bpoc), t-butoxycarbonyl (BOC), allyloxycarbonyl (Alloc), 9- fluorenylmethyloxycarbonyl (Fmoc), and benzyloxycarbonyl (Cbz); amide protecting groups, such as formyl, acetyl, trihaloacetyl, benzoyl, and nitrophenylacetyl; sulfonamide-protecting groups, such as 2-nitrobenzenesulfonyl; and imine and cyclic imide protecting groups, such as phthalimido and dithiasuccinoyl.
"Prodrug" is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound of the invention. Thus, the term "prodrug" refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable. A prodrug may be inactive when administered to a subject in need thereof, but is converted in vivo to an active compound of the invention. Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood. The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam)). A discussion of prodrugs is provided in Higuchi, T., et al., A. C. S. Symposium Series, Vol. 14, and in Bioreversible Carriers in Drug Design, Ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987. The term "prodrug" is also meant to include any covalently bonded carriers, which release the active compound of the invention in vivo when such prodrug is administered to a mammalian subject. Prodrugs of a compound of the invention may be prepared by modifying functional groups present in the compound of the invention in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound of the invention. Prodrugs include compounds of the invention wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the compound of the invention is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol or amide derivatives of amine functional groups in the compounds of the invention and the like.
The invention disclosed herein is also meant to encompass all pharmaceutically acceptable compounds of structure (I) being isotopically-labelled by having one or more atoms replaced by an atom having a different atomic mass or mass number. Examples of isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2H, 3H, 1 1C, 13C, 14C, 13N, 15N, 15O, 17O, 180, 31P, 32P, 35S, 18F, 36Cl, 123I, and 125I, respectively. These radiolabeled compounds could be useful to help determine or measure the effectiveness of the compounds, by characterizing, for example, the site or mode of action, or binding affinity to pharmacologically important site of action. Certain isotopically-labelled compounds of structure (I), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.
Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances. Substitution with positron emitting isotopes, such as 11C, 18F, 15O and 13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds of structure (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Preparations and Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
The invention disclosed herein is also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result from, for example, the oxidation, reduction, hydrolysis, amidation, esterification, and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising administering a compound of this invention to a mammal for a period of time sufficient to yield a metabolic product thereof. Such products are typically identified by administering a radiolabeled compound of the invention in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to human, allowing sufficient time for metabolism to occur, and isolating its conversion products from the urine, blood or other biological samples.
"Stable compound" and "stable structure" are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
"Mammal" includes humans and both domestic animals such as laboratory animals and household pets (e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like. "Optional" or "optionally" means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example, "optionally substituted aryl" means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution. "Pharmaceutically acceptable carrier, diluent or excipient" includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
"Pharmaceutically acceptable salt" includes both acid and base addition salts.
"Pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, camphoric acid, camphor- 10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane- 1 ,2-disulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, gluconic acid, glucuronic acid, glutamic acid, glutaric acid, 2-oxo-glutaric acid, glycerophosphoric acid, glycolic acid, hippuric acid, isobutyric acid, lactic acid, lactobionic acid, lauric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, mucic acid, naphthalene- 1 ,5-disulfonic acid, naphthalene-2-sulfonic acid, 1 -hydroxy-2-naphthoic acid, nicotinic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, propionic acid, pyroglutamic acid, pyruvic acid, salicylic acid, 4-aminosalicylic acid, sebacic acid, stearic acid, succinic acid, tartaric acid, thiocyanic acid, /?-toluenesulfonic acid, trifluoroacetic acid, undecylenic acid, and the like.
"Pharmaceutically acceptable base addition salt" refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol,
2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, 7V-ethylpiperidine, polyamine resins and the like. Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine. Often crystallizations produce a solvate of the compound of the invention. As used herein, the term "solvate" refers to an aggregate that comprises one or more molecules of a compound of the invention with one or more molecules of solvent. The solvent may be water, in which case the solvate may be a hydrate. Alternatively, the solvent may be an organic solvent. Thus, the compounds of the present invention may exist as a hydrate, including a monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding solvated forms. The compound of the invention may be true solvates, while in other cases, the compound of the invention may merely retain adventitious water or be a mixture of water plus some adventitious solvent. A "pharmaceutical composition" refers to a formulation of a compound of the invention and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans. Such a medium includes all pharmaceutically acceptable carriers, diluents or excipients therefor.
"Effective amount" or "therapeutically effective amount" refers to that amount of a compound of the invention which, when administered to a mammal, preferably a human, is sufficient to effect treatment, as defined below, of a bacterial infection in the mammal, preferably a human. The amount of a compound of the invention which constitutes a "therapeutically effective amount" will vary depending on the compound, the condition and its severity, the manner of administration, and the age of the mammal to be treated, but can be determined routinely by one of ordinary skill in the art having regard to his own knowledge and to this disclosure.
"Treating" or "treatment" as used herein covers the treatment of the disease or condition of interest in a mammal, preferably a human, having the disease or condition of interest, and includes:
(i) preventing the disease or condition from occurring in a mammal, in particular, when such mammal is predisposed to the condition but has not yet been diagnosed as having it;
(ii) inhibiting the disease or condition, i.e., arresting its development; (iii) relieving the disease or condition, i.e., causing regression of the disease or condition; or (iv) relieving the symptoms resulting from the disease or condition, i.e., relieving pain without addressing the underlying disease or condition. As used herein, the terms "disease" and "condition" may be used interchangeably or may be different in that the particular malady or condition may not have a known causative agent (so that etiology has not yet been worked out) and it is therefore not yet recognized as a disease but only as an undesirable condition or syndrome, wherein a more or less specific set of symptoms have been identified by clinicians.
The compounds of the invention, or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids. The present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (-), (R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). When the compounds described herein contain olefinic double bonds or other centres of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included.
A "stereoisomer" refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present invention contemplates various stereoisomers and mixtures thereof and includes "enantiomers", which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another.
A "tautomer" refers to a proton shift from one atom of a molecule to another atom of the same molecule. The present invention includes tautomers of any said compounds.
As noted above, in one embodiment of the present invention, compounds having antibacterial activity are provided, the compounds having the following structure (I):
Figure imgf000024_0001
(I) or a stereoisomer, pharmaceutically acceptable salt or prodrug thereof, wherein: wherein:
Q1 is hydroxyl, a protected hydroxyl, amino or a protected amino group; Q2 is hydrogen, optionally substituted alkyl,
Figure imgf000025_0001
Figure imgf000025_0002
Figure imgf000025_0003
Figure imgf000025_0004
Figure imgf000025_0005
Figure imgf000026_0001
Figure imgf000026_0002
Figure imgf000026_0003
each Ri and R2 is, independently, hydrogen or an amino protecting group; each R3 is, independently, hydrogen or a hydroxyl protecting group; each R4, R5, R7 and R8 is, independently, hydrogen or Ci -C6 alkyl optionally substituted with one or more halogen, hydroxyl or amino; each R6 is, independently, hydrogen, halogen, hydroxyl, amino or CpC6 alkyl; or R4 and R5 together with the atoms to which they are attached can form a heterocyclic ring having from 4 to 6 ring atoms, or R5 and one R6 together with the atoms to which they are attached can form a heterocyclic ring having from 4 to 6 ring atoms, or R4 and one R6 together with the atoms to which they are attached can form a carbocyclic ring having from 3 to 6 ring atoms, or R7 and R8 together with the atom to which they are attached can form a heterocyclic ring having from 4 to 6 ring atoms; each R9 is, independently, hydrogen, hydroxyl, amino or Ci-C6 alkyl; each Rio is, independently, hydrogen, halogen, hydroxyl, amino or Ci-C6 alkyl; or R9 and one Ri0 together with the atoms to which they are attached can form a heterocyclic ring having from 4 to 6 ring atoms; n is an integer from 0 to 4;
Zi and Z2 are both hydrogen or Zi and Z2 form a double bond; and
Z3 and Z4 are both hydrogen or Z3 and Z4 form a double bond.
In further embodiments, Q2 is hydrogen,
Figure imgf000027_0001
Figure imgf000028_0001
each R4, R5 and R6, independently, hydrogen or Ci-C6 alkyl, or R4 and R5 together with the atoms to which they are attached can form a heterocyclic ring having from 4 to 6 ring atoms, or R5 and R6 together with the atoms to which they are attached can form a heterocyclic ring having from 4 to 6 ring atoms, or R4 and R6 together with the atoms to which they are attached can form a carbocyclic ring having from 4 to 6 ring atoms.
In further embodiments, each Ri, R2 and R3 are hydrogen. In further embodiments, Qi is amino. In other further embodiments, Qi is hydroxyl.
In further embodiments, Zi and Z2 form a double bond. In other further embodiments, Zi and Z2 are both hydrogen.
In further embodiments, Z3 and Z4 form a double bond. In other further embodiments, Z3 and Z4 are both hydrogen. In further embodiments, Q2 is:
Figure imgf000028_0002
wherein: R4 is hydrogen; and R5 is hydrogen. In further embodiments, each R6 is hydrogen. For example, in more specific embodiments of the foregoing, Q2 is:
Figure imgf000028_0003
Figure imgf000029_0001
In other further embodiments, at least one R6 is halogen. In other further embodiments, Q2 is:
Figure imgf000029_0002
wherein: R4 is hydrogen; and R5 and one R6 together with the atoms to which they are attached form a heterocyclic ring having from 4 to 6 ring atoms. For example, in more specific embodiments of the foregoing, Q2 is:
Figure imgf000029_0003
In other further embodiments, at least one R6 is halogen. In other further embodiments, Q2 is:
Figure imgf000030_0001
wherein R4 and R5 together with the atoms to which they are attached form a heterocyclic ring having from 4 to 6 ring atoms. In further embodiments, each R6 is hydrogen. For example, in more specific embodiments of the foregoing, Q2 is:
Figure imgf000030_0002
Figure imgf000030_0003
In other further embodiments, at least one R6 is halogen. In other further embodiments, Q2 is:
Figure imgf000030_0004
wherein: R5 is hydrogen; and R4 and one R6 together with the atoms to which they are attached form a carbocyclic ring having from 3 to 6 ring atoms. For example, in more specific embodiments of the foregoing, Q2 is:
Figure imgf000031_0001
In other further embodiments, at least one R6 is halogen. In other further embodiments, Q2 is:
Figure imgf000031_0002
wherein: R4 is hydrogen; R7 is hydrogen; and R8 is hydrogen. In further embodiments, each R6 is hydrogen. For example, in more specific embodiments of the foregoing, Q2 is:
Figure imgf000031_0003
Figure imgf000032_0001
In other further embodiments, at least one R6 is halogen. In other further embodiments, Q2 is:
Figure imgf000032_0002
wherein: R4 and one R6 together with the atoms to which they are attached form a carbocyclic ring having from 3 to 6 ring atoms; R7 is hydrogen; and Rg is hydrogen. For example, in more specific embodiments of the foregoing, Q2 is:
Figure imgf000033_0001
In other further embodiments, at least one R6 is halogen. In other further embodiments, Q2 is:
Figure imgf000033_0002
wherein R5 is hydrogen. In further embodiments, each R6 is hydrogen. For example, in more specific embodiments of the foregoing, Q2 is:
Figure imgf000034_0001
In other further embodiments, at least one R6 is halogen. In other further embodiments, Q2 is:
Figure imgf000034_0002
wherein: R7 is hydrogen; and R8 is hydrogen. In further embodiments, each R6 is hydrogen. For example, in more specific embodiments of the foregoing, Q2 is:
Figure imgf000034_0003
In other further embodiments, at least one R6 is halogen. In other further embodiments, Q2 is:
Figure imgf000034_0004
wherein R5 is hydrogen. In further embodiments, each R6 is hydrogen. In other further embodiments, at least one R6 is halogen.
In other further embodiments, Q2 is:
Figure imgf000035_0001
wherein: R7 is hydrogen; and R8 is hydrogen. In further embodiments, each R6 is hydrogen. In other further embodiments, at least one R6 is halogen. In other further embodiments, Q2 is:
Figure imgf000035_0002
wherein R5 is hydrogen. In further embodiments, each R6 is hydrogen. In other further embodiments, at least one R6 is halogen.
In other further embodiments, Q2 is:
Figure imgf000035_0003
wherein R9 is hydrogen. In further embodiments, each Ri0 is hydrogen. In other further embodiments, at least one Ri0 is halogen.
In other further embodiments, Q2 is:
Figure imgf000036_0001
wherein: R7 is hydrogen; and R8 is hydrogen. In further embodiments, each R]0 is hydrogen. In other further embodiments, at least one R]0 is halogen. In other further embodiments, Q2 is:
Figure imgf000036_0002
wherein R4 is hydrogen. In further embodiments, each R6 is hydrogen. In other further embodiments, at least one R6 is halogen. In other further embodiments, Q2 is -C(=O)H.
In other further embodiments, Q2 is hydrogen. In other further embodiments, Q2 is optionally substituted alkyl. For example, in more specific embodiments of the foregoing, Q2 is unsubstituted or Q2 is substituted with one or more halogen, hydroxyl or amino.
In further embodiments, the foregoing compounds of structure (I) have the following configuration:
Figure imgf000037_0001
It is understood that any embodiment of the compounds of structure (I), as set forth above, and any specific substituent set forth herein for a Qi, Q2, Zi, Z2, Z3, Z4, Ri, R2, R3, R4, R5, R6, R7, R8, R9 and RJ 0 group in the compounds of structure (I), as set forth above, may be independently combined with other embodiments and/or substituents of compounds of structure (I) to form embodiments of the inventions not specifically set forth above. In addition, in the event that a list of substitutents is listed for any particular Qi, Q2, Z1, Z2, Z3, Z4, Ri, R2, R3 R4, R5, R6, R7, R8, R9 and R]0 group in a particular embodiment and/or claim, it is understood that each individual substituent may be deleted from the particular embodment and/or claim and that the remaining list of substituents will be considered to be within the scope of the invention.
For the purposes of administration, the compounds of the present invention may be administered as a raw chemical or may be formulated as pharmaceutical compositions. Pharmaceutical compositions of the present invention comprise a compound of structure (I) and a pharmaceutically acceptable carrier, diluent or excipient. The compound of structure (I) is present in the composition in an amount which is effective to treat a particular disease or condition of interest - that is, in an amount sufficient to treat a bacterial infection, and preferably with acceptable toxicity to the patient. The antibacterial activity of compounds of structure (I) can be determined by one skilled in the art, for example, as described in the Examples below. Appropriate concentrations and dosages can be readily determined by one skilled in the art.
The compounds of the present invention possess antibacterial activity against a wide spectrum of gram positive and gram negative bacteria, as well as enterobacteria and anaerobes. Representative susceptible organisms generally include those gram positive and gram negative, aerobic and anaerobic organisms whose growth can be inhibited by the compounds of the invention such as Staphylococcus, Lactobacillus, Streptococcus, Sarcina, Escherichia, Enterobacter, Klebsiella, Pseudomonas, Acinetobacter, Mycobacterium, Proteus, Campylobacter, Citrobacter, Nisseria, Baccillus, Bacteroides, Peptococcus, Clostridium, Salmonella, Shigella, Serratia, Haemophilus, Brucella and other organisms.
Administration of the compounds of the invention, or their pharmaceutically acceptable salts, in pure form or in an appropriate pharmaceutical composition, can be carried out via any of the accepted modes of administration of agents for serving similar utilities. The pharmaceutical compositions of the invention can be prepared by combining a compound of the invention with an appropriate pharmaceutically acceptable carrier, diluent or excipient, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols. Typical routes of administering such pharmaceutical compositions include, without limitation, oral, topical, transdermal, inhalation, parenteral, sublingual, buccal, rectal, vaginal, and intranasal. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques. Pharmaceutical compositions of the invention are formulated so as to allow the active ingredients contained therein to be bioavailable upon administration of the composition to a patient. Compositions that will be administered to a subject or patient take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of a compound of the invention in aerosol form may hold a plurality of dosage units. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 20th Edition (Philadelphia College of Pharmacy and Science, 2000). The composition to be administered will, in any event, contain a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, for treatment of a disease or condition of interest in accordance with the teachings of this invention. A pharmaceutical composition of the invention may be in the form of a solid or liquid. In one aspect, the carrier(s) are particulate, so that the compositions are, for example, in tablet or powder form. The carrier(s) may be liquid, with the compositions being, for example, an oral syrup, injectable liquid or an aerosol, which is useful in, for example, inhalatory administration. When intended for oral administration, the pharmaceutical composition is preferably in either solid or liquid form, where semi-solid, semi-liquid, suspension and gel forms are included within the forms considered herein as either solid or liquid.
As a solid composition for oral administration, the pharmaceutical composition may be formulated into a powder, granule, compressed tablet, pill, capsule, chewing gum, wafer or the like form. Such a solid composition will typically contain one or more inert diluents or edible carriers. In addition, one or more of the following may be present: binders such as carboxymethylcellulose, ethyl cellulose, microcrystalline cellulose, gum tragacanth or gelatin; excipients such as starch, lactose or dextrins, disintegrating agents such as alginic acid, sodium alginate, Primogel, corn starch and the like; lubricants such as magnesium stearate or Sterotex; glidants such as colloidal silicon dioxide; sweetening agents such as sucrose or saccharin; a flavoring agent such as peppermint, methyl salicylate or orange flavoring; and a coloring agent.
When the pharmaceutical composition is in the form of a capsule, for example, a gelatin capsule, it may contain, in addition to materials of the above type, a liquid carrier such as polyethylene glycol or oil.
The pharmaceutical composition may be in the form of a liquid, for example, an elixir, syrup, solution, emulsion or suspension. The liquid may be for oral administration or for delivery by injection, as two examples. When intended for oral administration, preferred composition contain, in addition to the present compounds, one or more of a sweetening agent, preservatives, dye/colorant and flavor enhancer. In a composition intended to be administered by injection, one or more of a surfactant, preservative, wetting agent, dispersing agent, suspending agent, buffer, stabilizer and isotonic agent may be included.
The liquid pharmaceutical compositions of the invention, whether they be solutions, suspensions or other like form, may include one or more of the following adjuvants: sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils such as synthetic mono or diglycerides which may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. Physiological saline is a preferred adjuvant. An injectable pharmaceutical composition is preferably sterile. A liquid pharmaceutical composition of the invention intended for either parenteral or oral administration should contain an amount of a compound of the invention such that a suitable dosage will be obtained.
The pharmaceutical composition of the invention may be intended for topical administration, in which case the carrier may suitably comprise a solution, emulsion, ointment or gel base. The base, for example, may comprise one or more of the following: petrolatum, lanolin, polyethylene glycols, bee wax, mineral oil, diluents such as water and alcohol, and emulsifϊers and stabilizers. Thickening agents may be present in a pharmaceutical composition for topical administration. If intended for transdermal administration, the composition may include a transdermal patch or iontophoresis device.
The pharmaceutical composition of the invention may be intended for rectal administration, in the form, for example, of a suppository, which will melt in the rectum and release the drug. The composition for rectal administration may contain an oleaginous base as a suitable nonirritating excipient. Such bases include, without limitation, lanolin, cocoa butter and polyethylene glycol. The pharmaceutical composition of the invention may include various materials, which modify the physical form of a solid or liquid dosage unit. For example, the composition may include materials that form a coating shell around the active ingredients. The materials that form the coating shell are typically inert, and may be selected from, for example, sugar, shellac, and other enteric coating agents. Alternatively, the active ingredients may be encased in a gelatin capsule.
The pharmaceutical composition of the invention in solid or liquid form may include an agent that binds to the compound of the invention and thereby assists in the delivery of the compound. Suitable agents that may act in this capacity include a monoclonal or polyclonal antibody, a protein or a liposome.
The pharmaceutical composition of the invention may consist of dosage units that can be administered as an aerosol. The term aerosol is used to denote a variety of systems ranging from those of colloidal nature to systems consisting of pressurized packages. Delivery may be by a liquefied or compressed gas or by a suitable pump system that dispenses the active ingredients. Aerosols of compounds of the invention may be delivered in single phase, bi-phasic, or tri-phasic systems in order to deliver the active ingredient(s). Delivery of the aerosol includes the necessary container, activators, valves, subcontainers, and the like, which together may form a kit. One skilled in the art, without undue experimentation may determine preferred aerosols. The pharmaceutical compositions of the invention may be prepared by methodology well known in the pharmaceutical art. For example, a pharmaceutical composition intended to be administered by injection can be prepared by combining a compound of the invention with sterile, distilled water so as to form a solution. A surfactant may be added to facilitate the formation of a homogeneous solution or suspension. Surfactants are compounds that non-covalently interact with the compound of the invention so as to facilitate dissolution or homogeneous suspension of the compound in the aqueous delivery system.
The compounds of the invention, or their pharmaceutically acceptable salts, are administered in a therapeutically effective amount, which will vary depending upon a variety of factors including the activity of the specific compound employed; the metabolic stability and length of action of the compound; the age, body weight, general health, sex, and diet of the patient; the mode and time of administration; the rate of excretion; the drug combination; the severity of the particular disorder or condition; and the subject undergoing therapy.
Compounds of the invention, or pharmaceutically acceptable derivatives thereof, may also be administered simultaneously with, prior to, or after administration of one or more other therapeutic agents. Such combination therapy includes administration of a single pharmaceutical dosage formulation which contains a compound of the invention and one or more additional active agents, as well as administration of the compound of the invention and each active agent in its own separate pharmaceutical dosage formulation. For example, a compound of the invention and the other active agent can be administered to the patient together in a single oral dosage composition such as a tablet or capsule, or each agent administered in separate oral dosage formulations. Where separate dosage formulations are used, the compounds of the invention and one or more additional active agents can be administered at essentially the same time, i.e., concurrently, or at separately staggered times, i.e., sequentially; combination therapy is understood to include all these regimens.
It is understood that in the present description, combinations of substituents and/or variables of the depicted formulae are permissible only if such contributions result in stable compounds.
It will also be appreciated by those skilled in the art that in the synthetic processes described herein the functional groups of intermediate compounds may need to be protected by suitable protecting groups. Such functional groups include hydroxy, amino, mercapto and carboxylic acid. As described above, suitable protecting groups for hydroxy include trialkylsilyl or diarylalkylsilyl (for example, f-butyldimethylsilyl, t- butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like, and suitable protecting groups for amino, amidino and guanidino include r-butoxycarbonyl, benzyloxycarbonyl, and the like. Suitable protecting groups for mercapto include -C(O)-R" (where R" is alkyl, aryl or arylalkyl), /?-methoxybenzyl, trityl and the like. Suitable protecting groups for carboxylic acid include alkyl, aryl or arylalkyl esters. Protecting groups may be added or removed in accordance with standard techniques, which are known to one skilled in the art and as described herein. The use of protecting groups is described in detail in Green, T.W. and P. G. M. Wutz, Protective Groups in Organic Synthesis (1999), 3rd Ed., Wiley. As one of skill in the art would appreciate, the protecting group may also be a polymer resin such as a Wang resin, Rink resin or a 2-chlorotrityl-chloride resin.
It will also be appreciated by those skilled in the art, although a protected derivative of compounds of this invention may not possess pharmacological activity as such, they may be administered to a mammal and thereafter metabolized in the body to form compounds of the invention which are pharmacologically active. Such derivatives may therefore be described as "prodrugs". All prodrugs of compounds of this invention are included within the scope of the invention.
Furthermore, all compounds of the invention which exist in free base or acid form can be converted to their pharmaceutically acceptable salts by treatment with the appropriate inorganic or organic base or acid by methods known to one skilled in the art. Salts of the compounds of the invention can be converted to their free base or acid form by standard techniques.
The following Examples illustrate various methods of making compounds of this invention, i.e., compound of structure (I):
Figure imgf000043_0001
(I) wherein Qi, Q2, Zi, Z2, Z3, Z4, Ri, R2 and R3 are as defined above. It is understood that one skilled in the art may be able to make these compounds by similar methods or by combining other methods known to one skilled in the art. It is also understood that one skilled in the art would be able to make, in a similar manner as described below, other compounds of structure (I) not specifically illustrated below by using the appropriate starting components and modifying the parameters of the synthesis as needed. In general, starting components may be obtained from sources such as Sigma Aldrich, Lancaster Synthesis, Inc., Maybridge, Matrix Scientific, TCI, and Fluorochem USA, etc. or synthesized according to sources known to those skilled in the art (see, for example, Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5th edition (Wiley, December 2000)) or prepared as described herein.
The following examples are provided for purposes of illustration, not limitation.
EXAMPLES
Example 1
Figure imgf000045_0001
To a stirring solution of (5)-4-(benzyloxycarbonylamino)-2- hydroxybutanoic acid (1, 612 g, 2.41 mol) in THF (6 L) at 0°C was added DMAP (2.4 g) and pyridine (196 mL, 2.41 mol). After stirring for 20 minutes, benzoyl chloride (280 mL) was added slowly and the reaction was allowed to stir overnight at room temperature. The resulting precipitate was filtered and the filtrate was acidified with 1 M citric acid (1 L). The organic solvent was removed by rotary evaporation and the desired product was extracted with EtOAc (2 x 2L). The combined organic layers were washed with 1 M citric acid (1 L), brine (1 L) and then dried over MgSO4. Solvent removal under vacuum resulted in a viscous oil, which was purified on a 6-inch reverse- phase HPLC column to yield (5)-2-(benzoyloxy)-4-(benzyloxycarbonylamino)butanoic acid (2) as a non-flowing viscous oil (439 g, 51% yield).
Figure imgf000045_0002
To a stirring solution of (5)-2-(benzoyloxy)-4- (benzyloxycarbonylamino)butanoic acid (2, 196 g, 0.548 mol) and pentafluorophenol
(111 g, 0.603 mol) in 1.3 L DMF at O0C was added EDC (126 g, 0.658 mol) and the reaction was stirred overnight. The reaction mixture was diluted with hexanes:EtOAc
(1.5 L, 1 :1) and with brine/water (1 L, 1 :1), and the organic layer was separated and washed with 1 M citric acid (3 x 600 mL), followed by sat. NaHCO3: IM NaOH (3 x 500 mL, 1 :1) and brine (500 mL). The organic layer was dried over MgSO4, filtered and concentrated under vacuum to yield (5)-4-(benzyloxycarbonylamino)-l-oxo-l-
(perfluorophenoxy)butan-2-yl benzoate (3, 255 g, 88% yield) as a white waxy solid that was used in the next step without further purification.
Figure imgf000046_0001
4 5
To a stirring solution of nitrophenol (4, 150 g, 1078 mmol) in ethyl acetate (2 L) at 0°C was added Cbz-CI (161.6 mL, 1132 mmol). DIPEA (221.5 mL, 1294 mmol) was then dripped into the reaction over 45 minutes. The reaction was allowed to warm to room temperature and was stirred for 30 minutes. The reaction was then cooled to 0°C and the precipitate was removed by filtration. The organic layer was washed with 1 M citric acid (1.5 L), 1 :1 NaHCO3:lM NaOH (1.5 L), and brine (1.5 L), dried over Na2SO4 and concentrated to a yellow solid (5, 298.8 g, 100% yield).
Figure imgf000046_0002
Paromomycin sulfate (6, 685 g free base, 1113 mmol) was dissolved in water (4 L) and then ACN (4 L) was slowly added to the reaction. The reaction was cooled to O0C and then DIPEA (1525 mL, 8910 mmol) was added. O-Cbz-2- nitrophenol (5, 304 g, 1113 mmol) was dissolved in ACN (1 L) and added to the reaction via an addition funnel in one hour. The reaction was stirred for 18 hours and then was concentrated to a red viscous liquid, which was dripped into ethanol (1 mL reaction mixture: 6 mL ethanol) to yield a pale yellow precipitate, which was collected by filtration, washed with ethanol and dried under high vacuum to yield a pale yellow solid (1073 g). MTO Dowex exchange resin (11.5 kg) was suspended in methanol (11.5 L). Next the yellow solid was added to the suspended resin, and the mixture was stirred for 3 hours. The resin was removed by filtration and washed with methanol (14 L). The organic layer was concentrated to 5 L and dripped into ethyl acetate (30 L) to yield 6"'-Cbz-paromomycin (7, 663 g, 79% yield) as a white solid.
Figure imgf000047_0001
To a stirring solution of 6"'-Cbz-paromomycin (7, 98.3 g, 131 mmol) in 850 mL DMF under N2 was added 160 mL DIPEA (918 mmol) and the flask was cooled to -5O0C (CO2/ACN). A solution of (5)-4-(benzyloxycarbonylamino)-l-oxo-l- (perfluorophenoxy)butan-2-yl benzoate (3, 50 g, 95 mmol) in 340 mL DMF was then added dropwise and the reaction progress was monitored by HPLC. Solvent evaporation under high vacuum yielded a viscous liquid, which was dissolved in ethyl acetate:IPA (500 mL, 3:2), and washed with water : acetic acid (800 ml :34 mL). The aqueous layer was extracted with ethyl acetate (400 mL), followed by EtOAciIPA (2 x 960 mL, 85:15). The combined organic layers were washed with 500 mL H2O (500 mL), and the combined aqueous layers were washed with EtOAc:IPA (800 mL 1 :1) and then with EtOAc:IPA (200 mL 1 :1). The combined organic layers were then concentrated under vacuum to 175 mL. This solution was diluted with 25 mL H2O and was purified on a 6-inch reverse phase HPLC column to yield (8) (35.6 g, 25% yield): MS: m/z (M+Na)+ calc: 1111.4, obs: 111 1.4.
Figure imgf000047_0002
6" '-Cbz- 1 -(iV-Cbz-4-amino-2(5)-benzoyloxy-butyryl)-paromomycin (8,
41.4 g, 28.93 mmol) was dissolved in DMF (300 mL), and the reaction was cooled to 0°C. 7V-(benzyloxycarbonyloxy)succinimide (28.84 g, 115.72 mmol) was then added, followed by DIPEA (40.3 mmol, 231.4 mmol) and the reaction was stirred for two hours and then quenched by the addition of 7V,N-dimethylaminopropylamine (4.59 mL, 36.34 mmol, 1.26 eq). The reaction was stirred for 1 hour and then was diluted with ethyl acetate (1.5 L). The organic layer was washed with 5% acetic acid: water (2 x IL), water (2 x IL), and brine (1 x IL), dried over Na2SO4 and concentrated to a white foam (9, 44.4 g, 103% yield). HCbz
Figure imgf000048_0001
Figure imgf000048_0002
To a stirring solution of 25.4 g (17.03 mmol) of 2',3,2'",6'"-tetra-Cbz-l- (7V-Cbz-4-amino-2(5)-benzoyloxy-butyryl)-paromomycin (9) in 100 niL benzaldehyde was added 1.0 mL TFA dropwise. After stirring for 5.5 hours, the reaction solution was dripped into a vigorously stirred solution of 1.7 L of 3:2 ether : hexanes. The resulting slurry was allowed to stir overnight. Filtration and solvent removal under vacuum gave a crude, which was purified on a 6-inch reverse-phase HPLC column to yield 6',4'- benzylidine-l-(7V-Cbz-4-amino-2(5)-benzoyloxy-butyryl)-per-Cbz-paromomycin (10, 12.5 g, 7.9 mmol, 46% yield): MS: m/z (M+Na)+ calc. 1601.59, obs. 1601.5.
Figure imgf000048_0003
6',4'-Benzylidine-l-(7V-Cbz-4-amino-2(5)-benzoyloxy-butyryl)-per-Cbz- paromomycin (10, 0.5 g, 0.317 mmol) was dissolved in pyridine (8 mL), and then
TBDPSCl (0.246 mL, 0.950 mmol, 3 eq) and DMAP (0.1 g, 0.079 mmol, 0.25 eq) were added to the reaction, which was heated at 8O0C for 18 hours. Solvent evaporation under vacuum gave a brown residue, which was purified on a 2-inch reverse-phase HPLC column to yield 6',4'-benzylidine-l-(7V-Cbz-4-amino-2(5)-benzoyloxy-butyryl)- per-Cbz-5"-OTBDPS-paromomycin (11, 0.220 g, 38% yield): MS: m/z (M+Na)+ calc. 1839.7, obs. 1840.6. HCbz
Figure imgf000049_0001
6',4'-Benzylidine-l-(7V-Cbz-4-amino-2(5)-benzoyl-butyryl)-per-Cbz-5"- OTBDPS-paromomycin (11, 0.187 g, 0.103 mmol) was dissolved in DCM (10 mL), placed under nitrogen, and cooled to 0°C. BH3-SMe2 in DCM (1 M, 0.545 mL, 1.545 mmol, 15 eq) was added and the reaction was stirred for two hours. The reaction was cooled to 0°C and BF3. Et2O (0.517 mL, 4.12 mmol, 40 eq) was added and the reaction was stirred. The reaction was quenched with NaHCO3, and diluted with DCM. The organic layer was separated, washed with NaHCO3, dried over Na2SO4, and concentrated to a white solid, which was purified by flash chromatography (silica gel, 0 to 10% MeOH/DCM) to yield 6'-benzyl-l-(N-Cbz-4-amino-2(5)-benzoyl-butyryl)-per- Cbz-5"-OTBDPS-paromomycin (12, 0.15 g, 80% yield). MS: m/z (M+Na)+ calc. 1841.72, obs. 1842.6. HCbz
Figure imgf000049_0002
6'-Benzyl-5"-TBDPS-l-(N-Cbz-4-amino-2(.Sr)-benzoyl-butyryl)-per- Cbz-paromomycin (12, 0.15 g, 0.0825 mmol) was dissolved in anhydrous toluene (6 mL) and anhydrous acetonitrile (1 mL). PPh3 (0.26 g, 0.99 mmol, 12 eq), triiodoimidazole (0.176 g, 0.396 mmol, 4.8 eq), and imidazole (0.034 g, 0.495 mmol, 6 eq) were added and the reaction was placed under nitrogen and heated to reflux for 4 hours and then allowed to cool to room temperature. The reaction was quenched by the addition of 5% by weight Na2S2O3 and stirred for 1 hour. The layers were then partitioned and the organic layer was concentrated to an orange oil, which was purified on a 2-inch reverse-phase HPLC column to yield 3',4',3'",4"'-tetradehydro-6'-benzyl- 5"-TBDPS-l-(N-Cbz-4-amino-2(5)-benzoyl-butyryl)-per-Cbz-paromomycin (13,
0.0734 g, 51% yield): MS: m/z (M+Na)+ calc. 1773.71, obs. 1774.51.
Figure imgf000050_0001
3 ' ,4' ,3 " ' ,4" ' -tetradehydro-6 ' -benzyl-5 " -TBDP S- 1 -(iV-Cbz-4-amino- 2(5)-benzoyl-butyryl)-per-Cbz-paromomycin (13, 0.073 g, 0.042 mmol) was dissolved in THF (2 mL). A solution of TBAF in THF (1 M, 0.42 mL, 0.42 mmol, 10 eq) was added and the reaction was placed under nitrogen and stirred for 8 hours. The reaction was quenched with NH4Cl and stirred for 10 minutes. The reaction was then diluted with ethyl acetate and the layers were partitioned. The organic layer was washed with NH4Cl, dried over Na2SO4, and concentrated to yield 3',4',3'",4'"-tetradehydro-6'- benzyl-1 -(N-Cbz-4-amino-2(5)-benzoyl-butyryl)-per-Cbz-paromomycin (14), which was used in the next step without further purification.
HCbz
Figure imgf000051_0001
3 ' ,4' ,3 " ' ,4" ' -Tetradehydro-6 ' -benzyl- 1 -(iV-Cbz-4-amino-2(5)-benzoyl- butyryl) -per-Cbz-paromomycin (14, 0.05 g, 0.033 mmol) was dissolved in MeOH (3 mL), methylamine (0.642 mL, 8.27 mmol, 250 eq) was added and the reaction was stirred for 6 hr. The reaction mixture was then diluted with ethyl acetate, washed with water, dried over Na2SO4 and concentrated to a yellow oil, which was purified on a 1- inch reverse-phase HPLC column to yield 3',4',3'",4"'-tetradehydro-6'-benzyl-l-(7V- Cbz-4-amino-2(,S)-hydroxy-butyryl)-per-Cbz-paromomycin (15): MS: m/z (M+Na)+ calc. 1431.56, obs. 1431.5.
NH2
Figure imgf000051_0002
3 ' ,4' ,3 " ' ,4" ' -Tetradehydro-6 ' -benzyl- 1 -(7V-Cbz-4-amino-2(5)-hydroxy- butyryl) -per-Cbz-paromomycin (15, 0.007 g, 0.004 mmol) was dissolved in acetic acid : water (3mL : 2mL), and then Pd(OH)2 (0.007 g) was added. The reaction was evacuated and replenished with hydrogen, and was stirred for 18 hours. The reaction was filtered and lyophilized to yield 3',4',3'",4'"-tetradeoxy-l-(4-amino-2(5)- hydroxy-butyryl)-paromomycin acetate salt (16, 2.7 mg, 84% yield): MS: m/z (M+Na)+ calc.675.36, obs.675.4; CLND: 95% purity. Example 2
Figure imgf000052_0001
To a stirring solution of paromomycin sulfate (1, 76 g, 84 mmol) in water (209 mL) and tetrahydrofuran (1084 mL) at 00C was added a solution of sodium carbonate in water (254 mL, 218 mmol, 0.86 M), followed by the dropwise addition of benzyl chloro formate (120 mL, 840 mmol). Sodium bicarbonate (70.6 g, 840 mmol) was then added and the reaction was stirred for 3 hours. The two layers were separated and the organic layer was concentrated to a thick oil, which was diluted with ethyl acetate (400 mL) to give a total volume of 1.2 L. This was then dripped into hexane (9
L), and a precipitate formed. The filtrate was decanted off and concentrated to a white foam (2) (69.85 g, 64.5% yield).
Figure imgf000052_0002
To a stirring solution of (2) (69.85 g, 0.054 mmol) in 200 mL benzaldehyde was added 2 mL TFA dropwise. After stirring for 5.5 hours, the reaction solution was dripped into a vigorously stirred solution of 3.4 L of 3:2 EtheπHexanes and the resulting slurry was allowed to stir overnight. Filtration and solvent removal under vacuum gave a crude, which was purified on a 6-inch reverse-phase HPLC column to yield 6',4'-benzylidine-per-Cbz-paromomycin (3, 35.7 g, 0.026 mmol, 48% yield).
Figure imgf000053_0001
6',4'-Benzylidine-per-Cbz-paromomycin (3) (1.98 g, 1.44 mmol) was dissolved in DCM (10 mL), placed under nitrogen, and cooled to 0°C. BH3-SMe2 in DCM (1 M, 14.6 mL, 14.56 mmol) was added and the reaction was stirred for two hours. Then the reaction was cooled to 0 0C and BF3.Et2O (1.46 mL, 11.64 mmol) was added and the reaction was stirred for one hour. The reaction was then diluted with
DCM, washed with NaHCO3, dried over Na2SO4, and concentrated to a crude, which was purified by flash chromatography (silica gel, DCM/MeOH) to yield a white solid (4, 0.756 g, 0.54 mmol, 37.5% yield).
TBDPSCI, DMAP pyridine
Figure imgf000053_0003
Figure imgf000053_0002
ό'-Benzyl-per-Cbz-paromomycin (4, 0.756 g, 0.54 mmol) was dissolved in pyridine (6 mL), and then TBDPSCl (0.429 mL, 1.65 mmol, 3 eq) and DMAP (0.067 g, 0.55 mmol, 1 eq) were added and the reaction was heated at 80°C for 18 hours. Solvent removal under vacuum gave a crude, which was purified on a 2-inch reverse- phase HPLC column to yield 6'-benzyl-5"-OTBDPS-per-Cbz-paromomycin (5, 0.167 g, 19% yield): MS: m/z (M+Na)+ calc.1636.64, obs.1636.6.
Figure imgf000054_0001
6'-Benzyl-5"-OTBDPS-per-Cbz-paromomycin (5, 2.8 g, 1.73 mmol) was dissolved in anhydrous toluene (170 mL) and anhydrous acetonitrile (40 mL). PPh3 (5.4 g, 20.8 mmol, 12.0 eq), triiodoimidazole (3.71 g, 8.3 mmol, 4.8 eq), and imidazole (0.709 g, 10.4 mmol, 6 eq.) were then added and the reaction was placed under nitrogen and refluxed for 4 hours and then allowed to cool to room temperature. The reaction was then quenched by the addition of aq. Na2S2O3 (400 mL of 0.3 M aqueous solution) and stirred for 1 hour. The layers were then partitioned and the organic layer was concentrated to an orange oil, which was purified on a 2-inch reverse- phase HPLC column to yield 3',4',3'",4'"-tetradehydro-6'-benzyl-5"-OTBDPS-per- Cbz-paromomycin (6, 1.46 g, 54.4% yield): MS: m/z (M+Na)+ calc.1568.63, obs.1569.1.
Figure imgf000054_0002
6'-Benzyl-5"-OTBDPS-3',4',3'",4'"-tetra-dehydro-per-Cbz- paromomycin (6, 0.2 g, 0.129 mmol) was dissolved in THF (2 mL). A solution of TBAF in THF (1 M, 1.29 mL, 1.29 mmol, 10 eq) was added and the reaction was placed under nitrogen and stirred for 8 hours. The reaction was quenched with NH4Cl and stirred for 10 minutes. The reaction was then diluted with ethyl acetate and the layers were partitioned. The organic layer was washed with NH4Cl, dried over Na2SO4, and concentrated to a brown oil, which was purified on 1-inch reverse-phase HPLC column to yield 6'-benzyl-3',4',3'",4'"-tetra-dehydro-per-Cbz-paromomycin (7) (0.057g, 0.044 mmol, 34% yield).
Figure imgf000055_0001
6'-Benzyl- 3',4',3'",4'"-tetra-dehydro-per-Cbz-paromomycin (7, 0.057 g, 0.044 mmol) was dissolved in acetic acid : water (3mL : 2mL), and then Pd(OH)2 (0.06 g) was added. The reaction was evacuated and replenished with hydrogen and was stirred for 18 hours. The reaction mixture was filtered, washed with water and lyophilized to yield 3',4',3'",4'"-tetra-deoxy-per-Cbz-paromomycin acetate salt (8, 7.7 mg, 32% yield): MS: m/z (M+Na)+ calc. 574.32, obs. 574.3; CLND 96.5% purity.
Example 3
Figure imgf000055_0002
Neomycin sulfate (1, 240 g, 391 mmol, 1 eq) was suspended in MeOH
(10 L) then was diluted with H2O (400 mL). The reaction was cooled to 0°C and K2CO3 (1080 g, 7818 mmol, 20 eq) was added slowly to the reaction mixture. Benzyl chloroformate (550 mL, 3910 mmol, 10 eq) was then added to the reaction mixture via a dropping funnel over 3 hours. The mixture was stirred vigorously for 18 hours at room temperature, then was filtered and concentrated to 2 L. The concentrate was diluted with ethyl acetate (2 L), washed with water (2 x 2L), washed with brine (2L), dried over Na2SO4 and concentrated to a yellow foam. The foam was dissolved in MeOH (1 L) and dripped into Et2O (24 L), and a precipitate was formed. The white solid was filtered off and dried under high vacuum to yield 410 g of crude, which was purified on a 6-inch reverse phase HPLC column to yield l,3,2',6',2'",6'"-hexa-Cbz- neomycin (2, 171 g, 31%). MS: m/z (M+Na)+ calc.1441.53, obs. 1441.6.
TBDPSCI, DMAP pyridine
Figure imgf000056_0002
Figure imgf000056_0001
l,3,2',6\2'",6'"-Hexa-Cbz-neomycin (2, 239 g, 169 mmol) was dissolved in pyridine (1.2 L) and then 4-dimethylaminopyridine (20.6 g, 168.6 mmol) was added. The reaction was heated at 80°C and then t-butyl-diphenylsilyl chloride (175.2 mL, 673.7 mmol) was added via addition funnel over one hour. The reaction was heated at 80°C and stirred for 18 hours and then was cooled to room temperature. The cooled reaction mixture (1.4 L) was dripped into vigorously stirred ether (30 L). The resultant precipitate was filtered and washed with ether (1 L). The precipitate was then dried under vacuum to yield 31O g of white solid, which was purified on a 6-inch reverse-phase HPLC column to yield l,3,2',6',2"\6"'-hexa-Cbz-5"-OTBDPS- neomycin (3, 238 g, 85 % yield). MS: m/z (M+Na)+ calc. 1679.65, obs. 1679.6.
Figure imgf000056_0003
l,3,2',6',2'",6'"-Hexa-Cbz-5"-OTBDPS-neomycin (3, 80 g, 48.3 mmol) was dissolved in anhydrous toluene (2.7 L) and acetonitrile (0.67 L). PPh3 (151.9 g, 580 mmol, 12.0 eq), triiodoimidazole (103.3 g, 232 mmol, 4.8 eq), and imidazole (19.73 g, 290 mmol, 6 eq.) were added and the reaction was placed under nitrogen and refluxed for 4 hours and then allowed to cool to room temperature. The reaction was then quenched by the addition of aq. Na2S2O3 (200 g in 1 L water) and stirred for 1 hour. The layers were then partitioned and the organic layer was concentrated to 600 mL. Then the reaction mixture was seeded with triphenyl phosphine oxide and placed in the freezer for 18 hours. The solids were removed by filtration, and the filtrate was concentrated to an orange-brown oil, which was purified on a 6-inch reverse-phase HPLC column to yield 3',4',3'",4'"-tetradehydro-l,3,2',6',2'",6'"-hexa-Cbz-5"- OTBDPS-neomycin (4, 50% yield): MS: m/z (M+Na)+ calc. 1611.64, obs. 1611.5.
Figure imgf000057_0001
3',4',3'",4'"-Tetra-dehydro-5"-TBDPS-l,3,2',6',2'",6'"-hexa-Cbz- neomycin (4, 96.3 g, 60.6 mmol) was dissolved in THF (90 mL). A solution of TBAF in THF (1 M, 606 mL, 606 mmol, 10 eq) was added and the reaction was placed under nitrogen and stirred for 4 hours. The reaction was quenched with NH4Cl (1.6 L) and stirred for 10 minutes. The reaction was then diluted with ethyl acetate (1.5 L) and the layers were partitioned. The organic layer was washed with NH4Cl (1.5 L), dried over Na2SO4, and concentrated to 400 mL. The organic layer (400 mL) was dripped into 2:1 ether:hexane (7.3L: 3.7L), and a precipitate was formed. The solids were collected by filtration and dried under vacuum to yield 3',4',3'",4'"-tetra-dehydro-l,3,2',6',2'",6'"- hexa-Cbz-neomycin (5, 78.24 g, 57.9 mmol, 94% yield): MS: m/z (M+Na)+ calc. 1373.52, obs. 1373.5.
Figure imgf000058_0001
To a solution of 500 mg of 3\4\3'",4'"-tetra-dehydro-l,3,2',6',2'",6'"- hexa-Cbz-neomycin (5, 0.37 mmol) in 10 mL acetic acid and 5 mL water was added
250 mg of Pd(OH)2 on carbon (20% dry basis Pd), and the reaction vessel was evacuated and replenished with hydrogen. The reaction was monitored by HPLC-MS and deemed complete after 1 hour. The reaction mixture was filtered through a .45 μm
PTFE filter and the filter was rinsed with an additional 5 mL water. The filtrate was diluted to a final volume of 100 mL with water and was then lyophilized to a crude, which was purified on a 1-inch reverse-phase HPLC column (0-10% ACN, 10 mM NH4OH) to yield 3',4',3'",4"'-tetra-deoxy-neomycin (6, 22.5 mg, 18% yield): MS: m/z
(M+Na)+ calc. 573.33, obs. 573.3; CLND 98% purity.
Example 4
Figure imgf000058_0002
3',4',3'",4'"-tetra-dehydro-6',2',3,l,2'",6'"-hexa-Cbz-neomycin (1, 78.24 g, 57.9 mmol) was dissolved in anhydrous DMA (1 L) and placed under nitrogen. In another round bottom flask NaH (6.26 g, 260.6 mmol, 4.5 eq) was suspended in anhydrous DMA (1 L), cooled to 0°C, and placed under nitrogen. The 3', 4', 3'", 4'"- tetra-dehydro-6',2',3,l,2"',6'"-hexa-Cbz-neomycin in DMA was added via dropping funnel into the cooled NaH suspension over 30 minutes. Then the reaction was stirred for two hours. Next the reaction was diluted with ethyl acetate (2 L), washed with water (2 x 2L), dried over Na2SO4, filtered, and concentrated to an orange oil, which was purified on a 6-inch reverse-phase HPLC column to yield 3',4',3'",4'"-tetra- dehydro-6',2',3,2'",6'"-penta-Cbz-l,6-oxazolidinone-neomycin as a white solid (2, 44.5 g, 62% yield): MS: m/z (M+Na)+ calc. 1265.46, obs. 1265.4.
Figure imgf000059_0001
To 44.5 g of 3',4\3'",4'"-tetra-dehydro-l,6-oxazolidinone- 3,2',6',2'",6'"-penta-Cbz-neomycin (2, 35.79 mmol) in 1.19 L of NMP was added 250 mL of 0.5 M aqueous LiOH (125.27 mmol, 3.5 eq) dropwise and the reaction was allowed to stir overnight. The reaction was quenched with the addition of 100 mL 1 M citric acid. The reaction mixture was further diluted with EtOAc (1 L) and brine (2 L), and the organic layer was separated. The aqueous layer was washed with 200 mL ethyl acetate and the combined organics were then washed with 600 mL of sat. aq. NaHCO3 followed by 600 mL of a 1 :1 mixture of water and brine. The combined organics were then washed with 600 mL of brine, dried over MgSO4, filtered and concentrated under vacuum to yield a cude, which was purified on a 6-inch reverse-phase HPLC column to yield 3',4',3'",4'"-tetra-dehydro-3,2',6',2'",6'"-penta-Cbz-neomycin as a white solid (3, 23.5 g, 65.6% yield): MS: m/z (M+Na)+ calc. 1239.49; obs. 1239.3.
Figure imgf000059_0002
To a solution of 21.6 g of 3',4',3'",4'"-tetra-dehydro-3,2',6',2'",6'"- penta-Cbz-neomycin (3, 17.75 mmol) in 800 mL of DMF at O0C and under a nitrogen atmosphere was added 11.25 g PyBOP (21.29 mmol, 1.2 eq) and 5.40 g of N-Cbz-4- amino-2(,S)-hydroxy-butanoic acid (21.29 mmol, 1.2 eq). The reaction flask was purged with nitrogen again and 4.56 mL of DIPEA (26.62 mmol, 1.5 eq) was added. The reaction was monitored by HPLC (214nm) and after 3 hours an extra 1.5 mL DIPEA was added (0.5 eq) and the reaction was stirred for 1 hr. The solution was neutralized by the addition of sat. aq. NH4Cl (100 mL). Citric acid (1 M, 100 mL) was then added, followed by brine (500 mL), and the aqueous layer was extracted with EtOAc (500 mL). The organic layer was washed with IM citric acid (500 mL), sat. aq. NaHCO3 (500 mL), brine (500 mL), dried over MgSO4, filtered and concentrated to dryness under high vacuum to yield a crude, which was purified on a 6-inch reverse- phase HPLC column to yield 3\4',3"',4"'-tetra-dehydro-3,2',6',2"',6"'-penta-Cbz-l- (N-Cbz-2(S)-hydroxy-butyryl)-neomycin as an off-white solid (4, 29.1 g, 85.5% yield): MS m/z (M+Na)+ calc. 1474.57; obs. 1474.3.
NH,
Figure imgf000060_0001
To a solution of 5.89 g of 3',4',3'",4'"-tetra-dehydro-3,2',6',2'",6'"- penta-Cbz-l-(N-Cbz-4-amino-2(1S)-hydroxy-butyryl)-neomycin (4, 0.091 mmol) in 200 mL acetic acid and 100 mL water was added 5.0 g of Pd(OH)2 on carbon (20% dry basis Pd), and the mixture was subjected to hydrogen at atmospheric pressure. The reaction was monitored by HPLC-MS and deemed complete after 1.5 hours. The reaction solution was filtered through Celite that had been rinsed with 200 mL water and the cake was rinsed with an additional 500 mL water. The filtrate was diluted to a final volume of 1.8 L with water and then lyophilized. The crude solid was purified on a 2-inch reverse-phase HPLC column (0-10% ACN, 10 niM NH4OH) to yield 3',4',3'",4'"-tetra-dehydro-l-(4-amino-2(5)-hydroxy-butyryl)-neomycin (5, 1.87 g, 74.8% yield): MS: m/z (M+Na)+ calc.674.38; obs. 674.4.
Example 5
Figure imgf000061_0001
To a solution of (1) (1.45 g, 1.022 mmol) in pyridine (19.3 mL) were added DMAP (cat.) and triphenylmethylene chloride (1.42 g, 5.111 mmol). The reaction mixture was stirred at 70 0C overnight. The solvent was evaporated under reduced pressure and the crude product was dissolved in CH2Cl2, washed with sat. NaHCO3, brine, dried (Na2SO4) and concentrated under reduced pressure. This crude material was purified by flash chromatography (Silica gel, DCM/MeOH) to give a white solid (2, 1.30 g, 77%). LRMS (ESI, m/z, M+H+) 1661.6; (M+NH4 +) 1678.7.
Figure imgf000061_0002
To a solution of (2) (1.30 g, 0.783 mmol) in toluene (43 mL) and MeCN
(10.75 mL) were added imidazole (0.320 g, 4.697 mmol), Ph3P (2.464 g, 9.394 mmol) and triiodoimidazole (1.675 g, 3.758 mmol). The solution was refluxed for 80 minutes, cooled and diluted with EtOAc. The organic layer was washed with 5% sodium thiosulfate, 0.5 N HCl, sat. NaHCO3, brine, dried (Na2SO4) and concentrated. The crude product was purified by flash chromatography (Silica gel, DCM/MeOH) to give a white solid (3, 0.810 g, 65%). LRMS (ESI, m/z, M+NH4 +) 1610.8.
Figure imgf000062_0001
A solution of (3) (0.810 g, 0.509 mmol) in dry DMF (17 mL) was cooled in an ice bath and to this solution was added sodium hydride (0.029 g, 1.221 mmol). The reaction mixture was stirred at 0 0C for 3 hours, quenched with aq. NH4Cl and then extracted with CH2Cl2. The combined organic layers were dried (Na2SO4) and concentrated in vacuo. The crude product was purified by flash chromatography (Silica gel, DCM/MeOH) to give a white solid (4, 0.464 g, 61%). LRMS (ESI, m/z, M+NH4 +) 1502.8.
,,NHCbZ
Figure imgf000062_0002
To a solution of (4) (0.394 g, 0.265 mmol) in DMF (15 mL) was added aq. LiOH (0.3 M, 2.65 mL, 0.796 mmol). The reaction mixture was stirred at room temperature for 6 hours. The reaction was quenched with aq. NH4Cl and extracted with CH2Cl2. The combined organic layers were dried (Na2SO4) and concentrated in vacuo. The crude product was purified by flash chromatography (Silica gel, DCM/MeOH) to give a white solid (5, 0.220 g, 57%). LRMS (ESI, m/z, M+H+) 1459.9. bz
,,NHCbZ
Figure imgf000063_0002
Figure imgf000063_0001
A solution of Cbz-HABA (0.095 g, 0.377 mmol), DCC (0.078 g, 0.377 mmol) and iV-hydroxysuccinimide (0.043 g, 0.377 mmol) in THF (10 mL) was stirred at room temperature for 30 min. To this solution were added (5) (0.220 g, 0.151 mmol) and NEt3 (126 μL, 0.905 mmol) and the reaction was allowed to stir at room temperature for an additional 2.5 hours. The solvent was then evaporated in vacuo and the concentrated crude product diluted with EtOAc, washed with 0.5 N HCl, sat.
NaHCO3, brine, dried (Na2SO4), filtered and concentrated under reduced pressure. The crude product was then purified by column chromatography (Silica gel, DCM/MeOH) to give a white solid (6, 0.210 g, 82%). LRMS (ESI) m/z (M+H)+ 1694.9; (M+NH4)+
1711.9.
Figure imgf000063_0003
Sodium metal (0.600 g), naphthalene (2.4 g) and tetrahydrofuran (20 mL) were combined under an inert atmosphere of nitrogen and stirred for 1 h. The solution became very dark green after 10 min. Using a syringe, this solution was added dropwise to a stirred solution of (6) (30 mg, 0.018 mmol) under nitrogen until the green color persisted (approx. 1 mL). After a few minutes the reaction turned from green to pink. Methanol (0.5 mL) was added, turning the reaction from pink to slightly yellow. The reaction was diluted with water (30 mL) then washed with diethyl ether (2 x 30 mL). The aqueous layer was acidified with acetic acid (0.5 mL) and washed with ethyl acetate (2 x 30 mL). The aqueous layer was frozen and lyophilized, then purified by preparative HPLC. The appropriate fractions were combined and the desired product was free-based with IRA-400 (OH form) ion-exchange resin, then filtered and lyophilized to yield (7) as a white solid (6 mg, 0.009 mmol, 50%). Calculated for C27H49N7On: 670.35 (M+Na)+; found: 670.3.
Example 6
Figure imgf000064_0001
Neomycin sulfate (1, 240 g, 391 mmol, 1 eq, mixture of B/C isomers) was suspended in methanol (10 L) then was diluted with water (400 mL). The reaction was cooled to O0C and K2CO3 (1080 g, 7818 mmol, 20 eq) was added slowly to the reaction mixture. Next benzyl chloro formate (550 mL, 3910 mmol, 10 eq) was added to the reaction mixture via a dropping funnel over 3 hours. The mixture was stirred vigorously for 18 hours at room temperature, then was filtered and concentrated to 2 L. The concentrate was diluted with ethyl acetate (2 L), washed with water (2 x 2 L) and brine (1 x 2 L), dried over Na2SO4 and concentrated to a yellow foam. The foam was dissolved in MeOH (1 L) and added dropwise into vigorously stirred Et2O (24 L). The white solids were filtered, washed with Et2O and dried under high vacuum to yield 410 g of crude product. This crude product was purified by reverse phase chromatography to yield the product as a white solid (2, 40 g, 7%). MS: m/z (M+Na)+ calc: 1441.53, obs: 1441.6. TBDPSCI, DMAP pyridine
Figure imgf000065_0002
Figure imgf000065_0001
Per-Z-neomycin C (2, 50.8 g, 35.8 mmol, 1 eq) was dissolved in pyridine (255 mL) and then 4-dimethylaminopyridine (4.36 g, 35.8 mmol, 1.0 eq) was added. The reaction was heated to 80 0C and then ^-butylchlorodiphenylsilane (36.63 mL, 143.2 mmol, 4.0 equiv.) was added via addition funnel over 30 minutes. The reaction was heated at 80 0C and stirred for 18 h and then was cooled to room temperature. The cooled reaction mixture was dripped into vigorously stirred ether (8.5
L). The resultant precipitate was filtered, washed with ether (1 L) and dried under vacuum to yield 50 g of white solid. The solid was purified by reverse phase chromatography to yield a white solid (3, 40.68 g, 68.6% yield). MS: m/z (M+Na) calc:
1679.65, obs: 1679.6.
Figure imgf000065_0003
5"-TBDPS-per-Cbz-Neomycin C (3, 40.15 g, 24.23 mmol, 1 eq) was dissolved in anhydrous toluene (1.34 L) and anhydrous acetonitrile (345 mL). Next
PPh3 (76.36 g, 291 mmol, 12.0 eq), triiodoimidazole (51.91 g, 116 mmol, 4.8 eq), and imidazole (9.91 g, 145.4 mmol, 6 eq.) were added to the reaction. The reaction was placed under nitrogen gas and heated to an internal temperature of 88 0C. The reaction was refluxed for 4 hours and then allowed to cool to room temperature. When the reaction solution had cooled to room temperature, the reaction was quenched by the addition OfNa2S2O3 (100 g in 1.6 L water) and stirred for 1 hour. The layers were then partitioned and the organic layer was concentrated to 400 mL. Then the reaction mixture was seeded with triphenylphosphine oxide and placed in the freezer for 18 hours. The solids were filtered, and the filtrate was concentrated to an orange-brown oil. The oil was purified by reverse phase column to yield a white solid (4, 17.82 g, 46% yield). MS: m/z (M+Na) calc: 1611.64, obs.: 1612.5
Figure imgf000066_0001
3',4',3'",4'"-tetra-dehydro-5"-TBDPS-per-Cbz-Neomycin C (4, 6.82 g, 4.29 mmol, 1 eq) was dissolved in THF (6.4 mL). Next IM TBAF in THF (43 mL, 42.9 mmol, 10 eq) was added to the reaction. The reaction was placed under nitrogen and stirred for 6 hours. Next the reaction was quenched with NH4Cl (115 mL) and stirred for 10 minutes. The reaction was then diluted with ethyl acetate (110 mL) and the layers were partitioned. The ethyl acetate layer was washed with NH4Cl (110 mL), dried over Na2SO4, and concentrated to 30 mL. The ethyl acetate (30 mL) was dripped into 2:1 etheπhexane (400 mL : 800 mL). The solids were filtered off and dried on a high vacuum line to yield a yellow solid (5, 5.69 g, 98% yield). MS: m/z (M+Na) calc: 1373.52, obs.: 1373.5.
Figure imgf000066_0002
3',4',3'",4'"-tetra-dehydro-per-Cbz-Neomycin C (5, 16.29 g, 12.1 mmol, 1 eq) was dissolved in anhydrous DMA (210 mL) and placed under nitrogen. In another round bottom flask NaH (1.6 g, 54.3 mmol, 4.5 eq) was suspended in anhydrous DMA (260 mL), cooled to 0 0C, and placed under nitrogen. The 3',4',3'",4'"-tetra-dehydro-per-Cbz-Neomycin in DMA was added via dropping funnel into the cooled NaH suspension over 30 minutes. Then the reaction was stirred for four hours. Next the reaction was diluted with ethyl acetate (500 mL), washed with water (2 x 500 mL), dried over Na2SO4, filtered, and concentrated to an orange oil. The oil was purified by reverse phase chromatography to yield a white solid (6, 8.18 g, 55% yield). MS: m/z (M+Na) calc: 1265.46, obs.: 1265.5.
ΛNHCbz
Figure imgf000067_0001
3',4',3'",4'"-tetra-dehydro-l ,6-oxazolidinone-per-Cbz-Neomycin C (6, 1 g, 0.8 mmol, 1 eq) was dissolved in NMP (27 mL) and then 0.5 M LiOH (0.117 g in 5.6 mL water, 2.8 mmol, 3.5 eq) was added via addition funnel over 30 minutes. The reaction was stirred for 18 hours and then was quenched with 1 M citric acid (100 mL) and the desired product was extracted into ethyl acetate (100 mL). The aqueous layer was back extracted with ethyl acetate (100 mL) and the combined organics were then washed with concentrated NaHCO3 (100 mL) followed by 1 :1 mixture of water and brine (100 mL). The organics were then dried over Na2SO4, filtered and concentrated to a yellow solid. The solid was purified by reverse phase chromatography to yield a white solid (7, 0.33 g, 34% yield). MS: m/z (M+Na) calc: 1239.46, obs.: 1239.5.
Figure imgf000067_0002
3',4',3'",4'"-tetra-dehydro-3,2',6',2'",6'"-Cbz-Neomycin (7, 0.331 g,
0.272 mmol, 1 eq) was dissolved in DMF (12 mL) and cooled to 0 0C. Next Cbz-(S)-
HABA (0.08 g, 0.33 mmol, 1.2 eq), PyBOP (0.170 g, 0.33 mmol, 1.2 eq), and DIPEA
(0.071 mL, 0.41 mmol, 1.5 eq) were added to the reaction. The reaction was stirred at 0 0C for 4 hours and then was diluted with ethyl acetate (50 mL). The organic layer was washed with H2O (50 mL), IM citric acid (50 mL), and NaHCO3 (50 mL), dried over
Na2SO4, filtered and concentrated to a white solid (0.3994 g). The solid was purified by reverse phase chromatography to yield a white solid (8, 0.2625 g, 66% yield). MS: m/z
(M+Na) calc: 1474.57, obs.: 1474.6.
Figure imgf000068_0001
3',4',3'",4'"-tetra-dehydro-3,2',6',2'",6'"-Cbz-l-[N-Cbz-(5)-HABA]- Neomycin (8, 0.2625 g 0.181 mmol, 1 eq) was dissolved in acetic acid (5 mL) and water (3 mL) and then Pd(OH)2 (0.2231 g) was added. The reaction was subjected to a hydrogen atmosphere. The reaction was stirred for 4 hours and then the palladium was filtered off and the reaction was dried down to a yellow-brown solid. The solid was purified by reverse phase column chromatography to yield a white solid (9, 0.0674 g, 57% yield). MS: m/z (M+H) calc: 652.38, obs.: 652.3.
Example 7
Figure imgf000069_0001
To 396 mg of (1) (3 from Example 4) (0.3253 mmol) dissolved in 4.0 mL DMF was added 122.5 μl DIPEA (0.7157 mmol) and the flask was cooled to O0C and purged with N2. The reaction was allowed to stir at room temperature overnight and progress was monitored by HPLC. When the reaction could be pushed no further, a few drops of water and TFA were added to bring the pH down to 2 and this solution was purified on a 2-inch RP-HPLC column to provide 241 mg pure (2) (54%). MS: m/z (M+Na) calc: 1379.46, obs: 1379.4.
Figure imgf000069_0002
To 127 mg of (2) (0.09314 mmol) dissolved in 2.1 mL DMF and under N2 was added 7.0 mg NaI (0.04657 mmol) and 7.2 mg NaN3 (0.1118 mmol). The reaction was then heated to 75 0C and the reaction progress was monitored by LCMS. After 7 hours, the reaction was deemed complete and was then quenched with the addition of an equal volume of water and product was extracted into 20 mL EtOAc. The organic layer was then washed twice with 50 mL water, once with brine, and dried over MgSO4. Solvent removal under high vacuum provided 110 mg of crude material which was purified by reverse-phase HPLC to provide 75 mg pure (3) (59% yield). MS: m/z (M+Na) calc: 1386.50, obs: 1386.3.
Figure imgf000070_0001
To 75 mg of (3) (0.05523 mmol) dissolved in 2 mL acetic acid and 700 μL water was added 75 mg Pd(OH)2 (20% Pd/C dry basis, 50% H2O) and the reaction was subjected to a hydrogen atmosphere and stirred for 2 hrs. The reaction solution was diluted with the addition of 10 mL H2O and was lyophilized. The crude material was purified by reverse-phase HPLC to obtain 15.8 mg of (4) (42.6% yield). MS: m/z (M+Na) calc: 694.35, obs: 694.4.
Example 8
Acetic Acid PyBOP DIPEA, DWIF
Figure imgf000070_0003
Figure imgf000070_0002
To a cold solution of 200 mg of 3',4\3'",4'"-tetra-dehydro- 3,2',6\2"',6"'-Cbz-neomycin (1 (3 from Example 4), 0.16 mmol) in 4.0 mL DMF was added 103 mg PyBOP (0.1972 mmol) and acetic acid (0.1972 mmol). The flask was then cooled to O0C and 47.8 μL DIPEA (0.2793 mmol) was added. The reaction was allowed to stir at room temperature overnight then LCMS was used to confirm product formation and the absence of starting material. The reaction was then quenched with 10-20 mL 1 M citric acid and desired product was extracted into 10 mL ethyl acetate. The organic layers were washed with 10 mL 1 M citric acid, brine and dried while stirring with MgSO4. Concentration under vacuum provided crude intermediate (2) that was used in the next reaction without further purification. MS: m/z (M+Na) calc: 1281.50, obs: 1281.4.
Figure imgf000071_0001
Crude (2) was dissolved in 40 niL 3:1 acetic acid:water and 190 mg
Pd(OH)2 (20% Pd dry basis, 50% H2O) was added and the reaction was subjected to a hydrogen atmosphere and stirred for 2 hrs. The reaction was filtered thru a 0.2 μm PTFE syringe filter then diluted with 120 mL water and lyophilized. The crude material was purified by reverse-phase HPLC to provide 54 mg pure (3) (55% yield, 2 steps). MS: m/z (M+H) calc: 593.34, obs: 593.3.
Example 9
Figure imgf000071_0002
To a cold solution of 200 mg of 3\4',3'",4'"-tetra-dehydro- 3,2',6',2'",6'"-Cbz-neomycin (1 (3 from Example 4), 0.16 mmol) in 4.0 mL DMF was added 103 mg PyBOP (0.1972 mmol) and 3-(tert-butoxycarbonylamino)-l- hydroxycyclobutanecarboxylic acid (0.1972 mmol). The flask was then cooled to 0 0C and 47.8 μL DIPEA (0.2793 mmol) was added. The reaction was allowed to stir at room temperature overnight then LCMS was used to confirm product formation and the absence of starting material. The reaction was then quenched with 10-20 mL 1 M citric acid and desired product was extracted into 10 mL ethyl acetate. The organic layers were washed with 10 mL IM citric acid, brine and dried while stirring with MgSO4. Concentration under vacuum provided crude intermediate (2) that was used in the next reaction without further purification. MS: m/z (M+Na) calc: 1452.59, obs: 1452.4.
Figure imgf000072_0001
Crude 2 was dissolved in 40 mL 3:1 acetic acid: water and 190 mg
Pd(OH)2 (20% Pd dry basis, 50% H2O) was added and the reaction was subjected to a hydrogen atmosphere and stirred for 2 hrs. The reaction was filtered thru a 0.2 μm PTFE syringe filter then diluted with 120 mL water and lyophilized. Crude (3) was used in the next reaction without further purification. MS: m/z (M+Na) calc: 786.43, obs: 786.4. H,
Figure imgf000072_0002
Crude (3) was dissolved in 200 μL water and the solution was cooled in an ice bath before 2 mL TFA were added. The reaction was monitored by LCMS and after 4 hours was complete. The reaction solution was diluted with 100 mL water and lyophilized. The resulting solids were purified by reverse-phase HPLC to provide 35.8 mg of (4) (0.0539 mmol, 33% over 2 steps). MS: m/z (M+Na) calc: 686.38, obs: 686.4.
Example 10
ΛNHCbz
Figure imgf000073_0002
Figure imgf000073_0001
To a cold solution of 200 mg of 3',4\3"',4"'-tetra-dehydro- 3,2',6',2'",6"'-Cbz-Neomycin (1 (3 from Example 4), 0.16 mmol) in 4.0 mL DMF was added 103 mg PyBOP (0.1972 mmol) and l-(tert-butoxycarbonyl)-3-hydroxyazetidine- 3-carboxylic acid (0.1972 mmol). The flask was then cooled to 0 0C and 47.8 μl DIPEA (0.2793 mmol) was added. The reaction was allowed to stir at room temperature overnight then LCMS was used to confirm product formation and the absence of starting material. The reaction was then quenched with 10-20 mL 1 M citric acid and desired product was extracted into 10 mL ethyl acetate. The organic layers were washed with a second 10 mL 1 M citric acid, brine and dried while stirring with MgSO4. Concentration under vacuum provided crude intermediate (2) that was used in the next reaction without further purification. MS: m/z (M+Na) calc: 1438.57, obs: 1438.4. C
Figure imgf000074_0001
Crude (2) was dissolved in 40 mL 3:1 acetic acid: water and 190 mg Pd(OH)2 (20% Pd dry basis, 50% H2O) was added and the reaction was subjected to a hydrogen atmosphere and stirred for 2 hrs. The reaction was filtered thru a 0.2 μm PTFE syringe filter then diluted with 120 mL water and lyophilized. Crude (3) was used in the next reaction without further purification. MS: m/z (M+Na) calc: 772.42, obs: 772.4.
Figure imgf000074_0002
Crude (3) was dissolved in 200 μL water and the solution was cooled in an ice bath before 2 mL TFA were added. The reaction was monitored by LCMS and after 4 hours was complete. The reaction solution was diluted with 100 mL water and lyophilized. The resulting solids were purified by reverse-phase HPLC column to provide 38 mg of (4) (0.0585 mmol, 35% over 2 steps). MS: m/z (M+Na) calc: 672.36, obs: 672.4.
General Synthetic Procedure for Examples 11-23
Procedure 1 : Hydro genolysis To a stirring solution of aminoglycoside (0.41 mmol) in AcOH (60 mL) was added H2O (15 mL), followed by Pd(OH)2/C (30% by weight, 50% H2O). The reaction vessel was evacuated and replenished with H2 (1 atm), and the reaction mixture was stirred for 4 hr. The solids were removed by filtration through a pad of Celite, and washed with H2O (100 mL). The aqueous layer was lyophilized to yield the desired product.
Example 11
Figure imgf000075_0001
To a stirring solution of 3',4',3'",4'"-tetra-dehydro-3,2',6',2'",6'"-penta- Cbz-neomycin (1, 200 mg, 0.164 mmol) in DMF (5 mL) was added 3-bromo- propionamide (0.192 mmol) followed by DIPEA (0.32 mmol), and a catalytic amount of NaI (0.016 mmol) and the reaction was heated to 40°C. The reaction progress was monitored by HPLC and in order to drive the reaction to completion, additional equivalents of 3-bromo-propionamide and DIPEA were added. A total of 5.5 equivalents of 3-bromo-propionamide (0.88 mmol) and 6.9 equivalents of DIPEA (1.10 mmol) were added, and after 6 days the reaction was deemed complete. The reaction mixture was purified on a 2-inch reverse-phase HPLC column to yield the desired 3',4',3'",4'"-tetra-dehydro-l-(3-propionamide)-3,2',6',2'",6'"-penta-Cbz-neomycin (2, 54 mg, 25.6% yield): MS: m/z (M+Na)+ calc. 1311.35, obs. 1311.4.
Figure imgf000076_0001
3',4',3'",4'"-Tetra-dehydro-l-(3-propionamide)-3,2',6',2'",6'"-penta- Cbz-neomycin (2, 54 mg, 0.042 mmol) was submitted to Procedure 1 for Cbz removal and reduction of the double bonds to give a crude, which was dissolved in cone. NH4OH (1.5 mL) and purified on a 1-inch reverse-phase HPLC column (0-10% ACN, 10 mM NH4OH) to yield 3\4',3"',4"'-tetra-deoxy-l-(3-propionamide)-neomycin (3, 16.0 mg, 0.026 mmol, 62% yield): MS: m/z (M+Na)+ calc. 644.37, obs.: 644.5; CLND 99.5% purity.
Example 12
Figure imgf000076_0002
To a stirring solution of 3',4',3"\4"'-tetra-dehydro-3,2',6\2"',6"'-penta- Cbz-neomycin (1, 205 mg, 0.168 mmol) in MeOH (4 mL) was added the epoxide (32.5 μL, 0.195 mmol), LiClO4 (45 mg, 0.42 mmol), and the reaction mixture was heated by microwave irradiation to 110°C for 7 hr. Additional epoxide (2 x 30 μL, 0.36 mmol) was added and the reaction was heated at 1100C for an additional 2 x 1 hr. The reaction mixture was then purified on a 2-inch reverse-phase HPLC column to yield 3',4',3'",4'"-tetra-dehydro-l-(N-Boc-2-hydroxy-3-amino-propyl)-3,2',6',2'",6'"-penta- Cbz-neomycin (2, 115 mg, 0.083 mmol, 49.4% yield): MS: m/z (M+Na)+ calc. 1412.59, obs.: 1412.4.
Figure imgf000077_0001
To a stirring solution of 3',4',3'",4'"-tetra-dehydro-l-(N-Boc-2- hydroxy-3-amino-propyl)-3,2',6',2'",6'"-penta-Cbz-neomycin (2, 84 mg, 0.060 mmol) in AcOH: H2O (4:1, 4 mL) was added Pd(OH)2 (65 mg), and the reaction mixture was stirred vigorously. The reaction was evacuated and replenished with a hydrogen atmosphere and was stirred for 2 hr. The reaction mixture was then filtered, and the solvent was concentrated to a residue. The residue was then dissolved in H2O (50 μL) and TFA (950 μL) was added and the reaction was stirred for 1 hr. The reaction was diluted with CH3CN (100 mL) and was concentrated to a crude, which was dissolved in cone. NH4OH and purified on a 1-inch reverse-phase HPLC column (0-10% ACN, 10 mM NH4OH) to yield 3',4',3'",4'"-tetra-deoxy-l-(2-hydroxy-3-amino-propyl)- neomycin (3, 5.8 mg, 0.0093 mmol, 15.5% yield): MS: m/z (M+Na)+ calc. 646.35, obs. 646.5; CLND 98.3% purity.
Example 13
Figure imgf000077_0002
To a stirring solution of 3',4\3'",4'"-tetra-dehydro-3,2',6',2'",6'"-penta- Cbz-neomycin (1, 200 mg, 0.164 mmol) in DMF (4 mL) was added N-Boc-2-hydroxy- 3-amino-propionic acid (47 mg, 0.197 mmol), followed by PyBOP (103 mg, 0.197 mmol) and DIPEA (0.048 mL, 0.279 mmol) and the reaction was stirred overnight. The 5 crude mixture was purified on a 2-inch reverse-phase HPLC column to yield 3 ' ,4' ,3 "',4"'-tetra-dehydro- 1 -(N-Boc-2(S)-hydroxy-3-amino-propionyl)- 3,2',6\2"',6"'-penta-Cbz-neomycin (2, 234 mg, 0.164 mmol, 99 % yield): MS m/z (M+Na)+ calc. 1426.57, obs. 1426.4.
Figure imgf000078_0001
3 ' ,4' ,3 ' ",4'"-Tetra-dehydro- 1 -(7V-Boc-2(S)-hydroxy-3 -amino- propionyl)- 3,2',6',2'",6'"-Cbz-neomycin (2, 234 mg, 0.164 mmol) was submitted to Procedure 1 for Cbz removal and reduction of the double bonds to give 3',4',3'",4'"- tetra-deoxy-l-(N-Boc-2(S)-hydroxy-3-amino-propionyl)-neomycin (3, 0.164 mmol): 15 MS: m/z (M+H)+ calc. 738.42, obs.738.3.
Figure imgf000078_0002
To a stirring solution of TFA (1 mL) and water (0.2 mL) at 00C was added 3',4',3'",4'"-tetra-deoxy-l-(N-Boc-2(5)-hydroxy-3-amino-propionyl)-neomycin
20 (3, 0.164 mmol) and the reaction was stirred for 30 minutes, and checked for completeness by MS. Upon completion, the reaction was diluted with water (10 mL) and lyophilized to yield a crude, which was dissolved in cone. NH4OH (1.5 mL) and purified on a 1-inch reverse-phase HPLC column (0-10% ACN, 10 mM NH4OH) to yield 3',4',3'",4'"-tetra-deoxy-l-(2(5)-hydroxy-3-amino-propionyl)-neomycin (4, 48.7 mg, 0.076 mmol, 40% yield): MS: m/z (M+H)+ calc. 638.0, obs.: 638.2 ; CLND 99.6% purity.
Example 14
Figure imgf000079_0001
To a stirring solution of 3',4',3"',4"'-tetra-dehydro-3,2\6\2"',6"'-penta- Cbz-neomycin (1, 200 mg, 0.16 mmol) in DMF (4 mL) was added 3-cyano-propionic acid (21 mg, 0.21 mmol), followed by PyBOP (120 mg, 0.22 mmol) and DIPEA (0.048 mL, 0.279 mmol) and the reaction was stirred overnight. The reaction mixture was then purified on a 2-inch reverse-phase HPLC column to yield 3',4',3'",4'"-tetra-dehydro-l- (3-cyano-propionyl)-3,2',6',2'",6'"-penta-Cbz-neomycin (105 mg, 51% yield): MS: m/z (M+Na)+ calc. 1320.5, obs. 1320.4.
Figure imgf000079_0002
3',4',3'",4'"-Tetra-dehydro-l-(3-cyano-propionyl)-3,2',6',2'",6'"-penta-
Cbz-neomycin (105 mg, 0.81 mmol) was submitted to Procedure 1 for Cbz removal and reduction of the double bonds to give a crude, which was dissolved in cone. NH4OH (1.5 mL) and purified on a 1-inch reverse-phase HPLC column (0-10% ACN,
10 mM NH4OH) to yield 3',4',3'",4'"-tetra-deoxy-l-(3-cyano-propionyl)-neomycin (3,
11 mg, 0.017 mmol, 21% yield): MS: m/z (M+Na)+ calc. 654.4, obs.654.4; CLND 94.4% purity; and 3',4',3'",4'"-tetra-deoxy-l-(3-amino-propionyl)-neomycin (4, 2 mg, 0.003 mmol, 4 %yield): MS: m/z (M+Na)+ calc. 658.4, obs. 658.4; CLND 91.9% purity.
Example 15
CbzHN
Figure imgf000080_0001
3',4',3'",4'"-Tetra-dehydro-3,2',6',2'",6'"-penta-Cbz-neomycin (1, 250 mg, 0.205 mmol) was dissolved in acetonitrile (1 mL) and concentrated to an oil, which was then dissolved in dry acetone (1 mL). Sodium carbonate (43.5 mg, 0.410 mmol) and iodoethanol (16.0 μL, 0.205 mmol) were added and the reaction was placed in a 350C heating block and stirred for 5 days and the reaction progress was monitored by HPLC. Additional portions of iodoethanol (2 x 2 μL) were added over the next 48 hr. Finally the reaction was quenched with sat. aq. ammonium chloride/water (2 mL, 10:1). The product was extracted with EtOAc (2 mL) and the combined organic layers were washed with sat. aq. NaHCO3 (1 mL), brine (1 mL), dried over MgSO4, filtered and concentrated to yield 3',4',3'",4'"-tetradehydro-l-(2-hydroxyethyl)-3,2',6',2'",6'"- penta-Cbz-neomycin (2, 0.205 mmol) as a white solid: MS: m/z (M+H)+ calc. 1261.5, obs.1261.3.
Figure imgf000081_0001
3',4',3'",4'"-Tetra-dehydro-l-(2-hydroxyethyl)-3,2',6',2'",6'"-penta- Cbz-neomycin (2, 0.041 mmol) was submitted to Procedure 1 for Cbz removal and reduction of the double bonds to give a crude, which was dissolved in cone. NH4OH (1.5 mL) and purified on a 1-inch reverse-phase HPLC column (0-10% ACN, 10 mM NH4OH) to yield 3',4',3'",4'"-tetra-deoxy-l-(2-hydroxyethyl)-neomycin (3, 9 mg, 0.015 mmol, 33% yield): MS: m/z (M+H)+ calc. 595.4, obs.595.3; CLND 99.3% purity.
Example 16
Figure imgf000081_0002
To a stirring solution of 3',4',3"',4"'-tetra-dehydro-3,2\6',2"\6"'-penta- Cbz-neomycin (1, 200 mg, 0.164 mmol) in MeOH (1 mL) was added 3- hydroxybenzaldehyde (20 mg, 0.164 mmol), followed by NaCNBH3 (12 mg, 0.19 mmol) and TFA (12 μl, 0.164 mmol) and the reaction was stirred overnight. To push the reaction to completion additional aldehyde and NaCNBH3 were added, and the reaction was stirred overnight. The reaction mixture was purified on a 2-inch reverse- phase HPLC column to yield 3',4',3'",4'"-tetra-dehydro-l-(3-hydroxybenzyl)- 3,2\6\2"',6"'-penta-Cbz-neomycin (2, 140 mg, 0.106 mmol, 66% yield): MS: m/z (M+H)+ calc. 1323.5, obs. 1323.3.
Figure imgf000082_0001
3 ' ,4' ,3 "\4 '"-Tetra-dehydro- 1 -(3 -hydroxybenzyl)-3 ,2 ' ,6 ' ,2 '",6 '"-penta- Cbz-neomycin (2, 140mg, 0.106 mmol) was submitted to Procedure 1 for Cbz removal and reduction of the double bonds to give a crude, which was dissolved in cone. NH4OH (1.5 mL) and purified on a 1-inch reverse-phase HPLC column (0-10% ACN, 10 niM NH4OH) to yield 3',4',3'",4'"-tetra-deoxy-l-(3-hydroxybenzyl)-neomycin (3, 56 mg, 0.085 mmol, 80% yield): MS: m/z (M+H)+ calc. 595.4, obs. 595.3; CLND 99.5% purity.
Example 17
CbzN VNHCbz
O y ' O
N y H
^NH2
QN HO-^^^ ^N^ .NHCbz
OH > OH NCbz
DIPEA, DMF, 80° C
1 2
To a stirring solution of 2(5)-hydroxy-4-amino-butyric acid (1, 0.059 g, 0.50 mmol) in DMF (2 ml) was added Λ/,7V'-bis(benzyloxycarbonyl)-lH-pyrazole-l- carboxamidine (0.26g, 0.70 mmol) followed by DIPEA (0.87 mL, 4.99 mmol) and the reaction was heated to 8O0C and stirred overnight. The crude mixture was purified on a 2-inch reverse-phase HPLC column to yield 7V,7V'-bis-Cbz-2(5)-hydroxy-4-guanidino- butyric acid (2): MS: m/z (M+H)+ calc. 430.15, obs. 430.1.
Figure imgf000083_0001
To a stirring solution of 3',4',3"',4"'-tetra-dehydro-3,2',6',2"',6"'-penta- Cbz-neomycin (2, 108 mg, 0.088 mmol) in DMF (1 mL) was added _V,_V'-bis-Cbz-2(_S)- hydroxy-4-guanidino-butyric acid (38 mg, 0.088 mmol) and the reaction was cooled to 0°C. PyBOP (50 mg, 0.097 mmol) was then added, followed by DIPEA (35 μL, 0.20 mmol) and the reaction was stirred. The reaction progress was monitored by HPLC and after 3 hr additional PyBOP (28 mg, 0.054 mmol) and DIPEA (18 μL, 0.10 mmol) were added and the reaction was stirred overnight. Additional PyBOP (38 mg, 0.073 mmol) and DIPEA (20 μL, 0.11 mmol) were added and the reaction was stirred overnight. The reaction mixture was then diluted with EtOAc (15 mL), and washed with 1 M citric acid (15 mL). The aqueous layer was back-extracted with EtOAc (15 mL). The combined organic layers were washed with 1 M citric acid (20 mL), sat. aq. NaHCO3 (15 mL), brine, dried over MgSO4, filtered and concentrated to a crude, which was purified on a 2-inch reverse-phase HPLC column to yield 3',4',3"',4"'-tetra-dehydro-l-(N,N'-bis- Cbz-2(5)-hydroxy-4-guanidino-butyryl)-3,2',6',2'",6'"-penta-Cbz-neomycin (3, 40 mg, 0.025 mmol, 28.4 % yield): MS: m/z (M+Na)+ calc. 1650.6, obs.1650.3.
Figure imgf000083_0002
3 ' ,4' ,3 '",4'"-Tetra-dehydro- 1 -(7V,iV'-bis-Cbz-2(1S)-hydroxy-4-guanidino- butyryl)- 3,2\6',2'",6"'-penta-Cbz-neomycin (3, 40 mg, 0.025 mmol) was submitted to Procedure 1 for Cbz removal and reduction of the double bonds to yield 3',4',3'",4'"- tetra-deoxy-l-(2(S)-hydroxy-4-guanidino-butyryl)-neomycin acetate salt (4, 23 mg, 0.021 mmol, 84% yield): MS: m/z (M+Na)+ calc. 716.4, obs.716.5; CLND 98.3% purity.
Example 18
Figure imgf000084_0001
To a stirring solution of 3',4\3"\4'"-tetra-dehydro-3,2\6\2'",6'"-penta- Cbz-neomycin (1, 200 mg, 0.164 mmol) in DMF (1 mL) was added glycidol (12.8 μL, 0.192 mmol) and the reaction was stirred at rt for 2 hr, and at 700C for 8 days. The reaction mixture was purified on a 2-inch reverse-phase HPLC column to yield 3',4',3'",4'"-tetra-dehydro-l-(2,3-bis-hydroxy-propyl)-3,2',6',2'",6'"-penta-Cbz- neomycin (2, 26 mg, 0.020 mmol, 13 % yield): MS: m/z (M+H)+ calc. 1291.5, obs.1291.3.
Figure imgf000084_0002
3',4',3'",4'"-Tetra-dehydro-l-(2,3-bis-hydroxy-propyl)-3,2',6',2'",6'"- penta-Cbz-neomycin (2, 26 mg, 0.020 mmol) was submitted to Procedure 1 for Cbz removal and reduction of the double bonds to yield 3',4',3'",4'"-tetra-deoxy-l-(2,3-bis- hydroxy-propyl)-neomycin acetate salt (3, 8.5 mg, 0.0087 mmol, 43.5% yield): MS: m/z (M+Na)+ calc. 647.4, obs. 647.5; CLND 96.8% purity. Example 19
DMF
Figure imgf000085_0002
Figure imgf000085_0001
To a stirring solution of 3\4',3"',4"'-tetra-dehydro-3,2',6',2"',6"'-penta- Cbz-neomycin (1, 200 mg, 0.164 mmol) in DMF (4 mL) was added JV-Cbz-glycine (41 mg, 0.197 mmol) and the reaction was cooled to 00C. PyBOP (103 mg, 0.197 mmol) and DIPEA (0.049 mL, 0.28 mmol) were added and the reaction was stirred for 3 hr. The reaction mixture was purified on a 2-inch reverse-phase HPLC column to yield 3',4',3'",4'"-tetra-dehydro-l-(N-Cbz-2-amino-acetyl)-3,2',6',2'",6'"-penta-Cbz- neomycin (2, 220 mg, 0.156 mmol, 95 % yield): MS: m/z (M+Na)+ calc. 1430.54, obs. 1430.4.
Figure imgf000085_0003
3',4',3'",4'"-Tetra-dehydro-l-(N-Cbz-2-amino-acetyl)-3,2',6',2'",6'"- penta-Cbz-neomycin (2, 220 mg, 0.156 mmol) was submitted to Procedure 1 for Cbz removal and reduction of the double bonds to give a crude, which was dissolved in cone. NH4OH (1.5 mL) and purified on a 1-inch reverse-phase HPLC column (0-10% ACN, 10 mM NH4OH) to yield 3',4',3'",4'"-tetra-deoxy-l-(2-aminoacetyl)-neomycin (3, 46 mg, 0.076 mmol, 48.7% yield): MS: m/z (M+H)+ calc. 608.69, obs. 608.3; CLND 99% purity.
Example 20
Figure imgf000086_0001
To a stirring solution of 3',4',3"',4"'-tetra-dehydro-3,2\6\2"',6"'-penta- Cbz-neomycin (1, 200 mg, 0.164 mmol) in DMF (4 mL) was added 7V-Cbz-L-serine (47 mg, 0.197 mmol) and the reaction was cooled to 00C. PyBOP (103 mg, 0.197 mmol) and DIPEA (0.049 mL, 0.28 mmol) were added and the reaction was stirred overnight. The reaction mixture was purified on a 2-inch reverse-phase HPLC column to yield 3',4',3'",4'"-tetra-dehydro-l-(iV-Cbz-2((S)-ammo-3-hydroxy-propionyl)- 3,2',6',2'",6'"- penta-Cbz-neomycin (2, 175 mg, 0.122 mmol, 74.4 % yield): MS: m/z (M+Na)+ calc. 1460.55, obs.1460.5.
Figure imgf000086_0002
3',4',3'",4'"-Tetra-dehydro-l-(N-Cbz-2(5)-amino-3-hydroxy- propionyl)-3,2',6',2'",6'"-penta-Cbz-neomycin (2, 175 mg, 0.122 mmol) was submitted to Procedure 1 for Cbz removal and reduction of the double bonds to give a crude, which was dissolved in cone. NH4OH (1.5 mL) and purified on a 1-inch reverse-phase HPLC column (0-10% ACN, 10 mM NH4OH) to yield 3\4',3'",4'"-tetra-deoxy-l- (2(S)-amino-3-hydroxy-propionyl)-neomycin (3, 47.5 mg, 0.075 mmol, 61.4% yield): MS: m/z (M+Na)+ calc. 660.72, obs. 660.4; CLND >99% purity.
Example 21
Figure imgf000087_0001
To a stirring solution of 3',4',3'",4'"-tetra-dehydro-3,2',6',2'",6'"-penta- Cbz-neomycin (1, 200 mg, 0.164 mmol) in MeOH (2 mL) was added NaCNBH3 (20 mg, 0.32 mmol), followed by l,3-dihydroxypropan-2-one (17.3 mg, 0.096 mmol) and the reaction was stirred for 5 hr. Additional NaCNBH3 (10 mg, 0.164 mmol) and 1 ,3- dihydroxypropan-2-one (8.6 mg, 0.048 mmol) were added and the reaction was stirred. The reaction was then diluted with EtOAc (10 mL), and the aqueous phase was separated. The organic layer was washed with sat. aq. NaHCO3 (10 mL). The aqueous layers were back-extracted with EtOAc (10 mL). The combined organic layers were washed with sat. aq. NaHCO3 (10 mL), sat. aq. NH4Cl (10 mL), brine (10 mL), dried over Na2SO4, filtered and concentrated to give a crude, which was purified on a 2-inch reverse-phase HPLC column to yield 3',4',3'",4'"-tetra-dehydro-l-(2-hydroxy-l- hydroxymethyl-ethyl)-3,2',6',2'",6'"-penta-Cbz-neomycin (2, 56 mg, 0.043 mmol, 26.2 % yield).
Figure imgf000088_0001
3 ' ,4' ,3 '",4'"-Tetra-dehydro- 1 -(2 -hydroxy- 1 -hydroxymethyl-ethyl)- 3,2',6',2'",6'"-penta-Cbz-neomycin (2, 56 mg, 0.043 mmol) was submitted to Procedure 1 for Cbz removal and reduction of the double bonds to give a crude, which was dissolved in cone. NH4OH (1.5 mL) and purified on a 1-inch reverse-phase HPLC column (0-10% ACN, 10 mM NH4OH) to yield 3',4',3'",4'"-tetra-deoxy-l-(2-hydroxy- l-hydroxymethyl-ethyl)-neomycin (3, 14 mg, 0.022 mmol, 51.2% yield): MS: m/z (M+Na)+ calc. 647.7, obs. 647.5; CLND 99.4% purity.
Example 22
Figure imgf000088_0002
NaHCO3, NaOH, DCM
1 2
To a solution of benzyl-iV-(2-aminoethyl)carbamate chloride salt (1, 540 mg, 2.34 mmol) in sat. aq. NaHCO3 (45 mL) was added 1 M NaOH (15 mL) and the reaction was stirred vigorously. DCM (30 mL) was added, followed by benzoylperoxide (1.13 g, 4.68 mmol) and the reaction was stirred overnight. The organic layer was separated and washed with brine, dried over MgSO4, filtered and concentrated to a crude, which was purified on a 1-inch reverse-phase HPLC column to yield benzyl-2-(benzoyloxyamino)ethyl carbamate (2, 252 mg, 0.80 mmol, 34.2%): MS: m/z (M+H)+ calc. 315.13, obs. 315.0.
Figure imgf000089_0001
3
To a stirring solution of disuccinimidyl carbonate (525 mg, 2.05 mmol) in CH3CN (16 mL) was added benzyl-2-(benzoyloxyamino)ethyl carbamate (2, 252 mg, 0.80 mmol) as a solution in CH3CN (12 mL) over 4 hours, and the reaction was stirred overnight. Additional disuccinimidyl carbonate (251 mg, 0.98 mmol) was added and the reaction was heated at 60°C overnight. Solvent removal gave a crude, which was purified on a 2-inch reverse-phase HPLC column to yield succinimidyl benzoyloxy(2- Cbz-aminoethyl)carbamate (3, 81 mg, 0.18 mmol, 22.5% yield).
Figure imgf000089_0002
Cbz-neomycin (4, 190 mg, 0.156 mmol) in DMF (2 mL) was added succinimidyl benzoyloxy(2-Cbz-aminoethyl)carbamate (3, 81 mg, 0.18 mmol) and the reaction was stirred under a nitrogen atmosphere. DIPEA (67 μL, 0.38 mmol) was added and the reaction was stirred for 2 days. The reaction mixture was purified on a 2-inch reverse- phase HPLC column, followed by treatment with sat. aq. NaHCO3 to remove the benzoyl protecting group to yield a crude, which was purified on a 1-inch reverse-phase column to yield (5) (60 mg, 0.041 mmol, 26.3 % yield): MS m/z (M+Na)+ calc. 1475.5, obs.1475.4.
Figure imgf000090_0001
Compound (5) (60 mg, 0.041 mmol) was submitted to Procedure 1 for
Cbz removal and reduction of the double bonds to give a crude, which was dissolved in cone. NH4OH (1.5 mL) and purified on a 1-inch reverse-phase HPLC column (0-10% ACN, 10 mM NH4OH) to yield (6) (10 mg, 0.015 mmol, 36.6% yield): MS: m/z
(M+H)+ calc. 653.7, obs.653.2; CLND 97.5% purity.
Example 23
Figure imgf000090_0002
To a stirring solution of 3',4',3'",4'"-tetra-dehydro-3,2',6',2'",6'"-penta- Cbz-neomycin (1, 397 mg, 0.326 mmol) in DCM (4 mL) was added DIPEA (223 μL, 1.30 mmol), followed by 2-phthalimido-ethane sulfonyl chloride (107 mg, 0.39 mmol) and the reaction was stirred overnight. Additional sulfonyl chloride (50 mg, 0.18 mmol) was added and the reaction was stirred overnight. Solvent removal gave a crude, which was dissolved in EtOAc and washed with H2O (2 x), brine, dried over MgSO4, filtered and concentrated to dryness to yield (2) (420 mg, 0.288 mmol, 88.3% yield): MS m/z (M+Na)+ calc. 1476.49, obs.1476.3.
Figure imgf000091_0001
To a stirring solution of (2) (420 mg, 0.288 mmol) in MeOH (4 mL) was added hydrazine (27.2 μL, 0.87 mmol) and the reaction was stirred overnight. The organic layer was washed with H2O (2 x), brine, dried over MgSO4, filtered and concentrated to a crude, which was purified on a 2-inch reverse-phase HPLC column to yield 3 (129 mg, 0.097 mmol, 33.7% yield): MS m/z (M+Na)+ calc. 1346.5, obs.1346.4.
Figure imgf000091_0002
3 ' ,4' ,3 '",4 ' "-Tetra-dehydro- 1 -(2-aminoethylsulfonamide)- 3,2',6',2'",6'"-penta-Cbz-neomycin (3, 129 mg, 0.097 mmol) was submitted to Procedure 1 for Cbz removal and reduction of the double bonds to give a crude, which was dissolved in cone. NH4OH (1.5 mL) and purified on a 1-inch reverse-phase HPLC column (0-10% ACN, 10 mM NH4OH) to yield 3',4\3"',4"'-tetra-deoxy-l-(2-amino- ethylsulfonamide)-neomycin (4, 34.7 mg, 0.053 mmol, 54.6 % yield): MS: m/z (M+Na)+ calc. 680.34, obs.680.4; CLND 99% purity.
General Synthetic Procedures for Examples 24-29
Procedure 2: Ozonolysis and Pinnick oxidation The substrate olefin (0.5 to 0.75 mmol) was dissolved in DCM (30 rnL) and the reaction was cooled to -78°C. Ozone was bubbled through until a blue color persisted (3 to 5 min), and the reaction was stirred for 1 hr. Argon was then bubbled through to remove excess ozone for 10 minutes. The reaction was further quenched by the addition of dimethyl sulfide (10 equiv.), and was stirred for 30 min with warming to rt. The solvent was reduced under vacuum to yield the crude aldehyde, which was dried under high- vacuum for 10 min, and used without further purification. To a stirring solution of the aldehyde in THF, ΦuOH and H2O (3:3:2, 10 mL), was added NaH2PO4 (4 equiv.) followed by 2-methyl-2-butene (10 equiv.) and sodium chlorite (2 equiv.), and the reaction was stirred for 4 hr. The reaction mixture was then added to sat. aq. NaCl (10 mL) and extracted with DCM (3x). The combined organic layers were dried over Na2SO4, filtered and reduced under vacuum to yield a crude, which was purified by flash chromatography (silica gel, 0 → 0.5 or 1% MeOH/DCM).
Procedure 3 : Amide coupling
To a stirring solution of the acid (0.060 mmol) and TV- hydroxysuccinimide (7.4 mg, 0.063 mmol) in EtOAc (2 mL) was added N,N'- dicyclohexylcarbodiimide (12.4 mg, 0.060 mmol) and the reaction mixture was stirred for 1 hr. The resulting white suspension was filtered through cotton, washed with EtOAc, and evaporated to dryness under vacuum to yield the activated ester. To a stirring solution of the activated ester in THF (2 mL) was added NaHCO3 (15 mg, 0.18 mmol) followed by 3',4',3'",4'"-tetra-dehydro-3,2',6',2'",6'"-Cbz-neomycin (50 mg, 0.041 mmol), and the reaction was stirred for 24 hr. The reaction mixture was quenched with sat. aq. NaHCO3 and extracted with DCM (3 x). The combined organic layers were dried over Na2SO4, filtered and reduced under vacuum to yield a crude, which was purified by flash chromatography (silica gel, 1 — > 1.5% MeOH/DCM) to yield the desired products as white amorphous solids.
Procedure 4: Hvdrogenolysis To a stirring solution of aminoglycoside (0.2 to 0.4 mmol) in THF (0.5 mL), was added MeOH (2 mL), followed by AcOH (20 μL), and the reaction mixture was stirred vigorously. Palladium hydroxide (50 mg, 20% wt) was then added and the reaction was evacuated and replenished with H2 (1 atmosphere); after 2 hr, water (2 mL) was added and stirring was continued overnight. The black suspension was filtered through a 0.45 μm syringe filter, neutralized with 28% aq. NH4OH and lyophilized. The resulting residue was dissolved in CHCl3: MeOH: NH4OH (2:3:1) and purified by flash chromatography (silica gel, CHCl3:MeOH:NH4OH, 2:3:1, 10 → 30% NH4OH). The fractions containing the desired product (R/= 0.1, CHCl3:MeOH:NH4OH, 2:3:2,) were collected, reduced under vacuum to yield a residue, which was dissolved in water, filtered through a 0.45 μm syringe filter, treated with AcOH (20 μl) and lyophilized to yield the desired aminoglycosides as their acetate salt.
Example 24
OH
Figure imgf000093_0001
Molecular sieves (4 A, 4 g) were added to a round bottom flask, and were activated by heating with a Bunsen burner under high vacuum. DCM (100 mL) was then added and the flask was cooled to -35°C with a cryocooler. Titanium tetraisopropoxide (1.75 mL, 5.95 mmol) and (i?,i?)-(-)-diisopropyl tartrate (1.65 mL, 7.75 mmol) were added and the reaction was stirred for 30 min. Penta-l,4-dienol (5 g, 59.4 mmol) and excess cumene hydroperoxide (80%, 17.5 mL) were added in small portions, and stirring was continued at -35°C for 48 hr. The reaction was quenched by addition of sat. aq. Na2SO4 (5 mL) immediately followed by Et2O (50 mL) and the reaction was stirred for 2 hr with warming to rt. The reaction mixture was filtered through Celite, and washed with Et2O. Solvent removal under vacuum without heating resulted in approximately 30 mL of a yellow solution. Excess cumene alcohol and hydroperoxide were removed by flash chromatography (silica gel, 40% Et2O/hex). Finally solvent removal under vacuum without heating yielded a mixture of (2S, 3R)- l,2-epoxy-4-penten-3-ol (1) (R/= 0.47, 1 :1 EtO Ac/hex) and diisopropyl tartrate (R/= 0.6), which was used in the next step without further purification. To a stirring solution of epoxide (1) in THF (100 mL) under an argon atmosphere was added tetrabutyl ammonium iodide (2.2 g, 5.96 mmol), followed by benzyl bromide (8.6 mL, 71.9 mmol) and the reaction was cooled to -15°C. Sodium hydride (60% in mineral oil, 2.65 g, 66.1 mmol) was added in small portions and the reaction was stirred overnight with warming to it. The reaction was quenched with MeOH, filtered through Celite, and washed with Et2O. Solvent removal gave an oily residue which was purified by flash chromatography (silica gel, 5 → 10% Et2O/hex) to yield (2S, 3i?)-l,2-epoxy-3-benzyloxy-4-pentene (2) as a clear non-volatile liquid (5.3g, 47.6% yield): R/= 0.69 (1 :4 EtO Ac/hex); [α]D = -36.7° (c 1.52, CHCl3); HRMS (ESI) (M+H)+ calc. for C12H14O2 191.1067, obs. 191.1064; 1H NMR (CDCl3, 300 MHz) δ 7.38-7.33 (m, 5H), 5.92-5.78 (m, IH), 5.41-5.39 (m, IH), 5.37-5.33 (m, IH), 4.66 (d, J = 11.95 Hz, lH), 4.49 (d, J = 11.96 Hz, IH), 3.83 (dd, J = 7.34, 4.20 Hz, IH), 3.10 (dt, J = 4.07, 4.06, 2.70 Hz, IH), 2.79 (dd, J = 5.21, 4.00 Hz, IH), 2.70 (dd, J = 5.23, 2.64 Hz, IH). 13C NMR (CDCl3, 100 MHz) δ 138.32, 134.67, 128.56 (2C), 127.87 (2C), 127.82, 119.73, 79.54, 70.83, 53.41, 45.00.
Figure imgf000094_0001
2 3
NaN3 (3.38 g, 52 mmol) and NH4Cl (2.78 g, 52 mmmol) in H2O (10 mL) were heated until a clear solution was obtained. This solution was then added dropwise to a solution of (25, 37?)-l,2-epoxy-3-benzyloxy-4-pentene (2) (3.3 g, 17.4 mmol) in MeOH (200 mL) and the reaction mixture was stirred for 4 days. The organic solvent was removed under vacuum, and the aqueous layer was extracted with DCM (3 x). The combined organic layers were dried over Na2SO4, filtered and reduced under vacuum to yield a crude, which was purified by flash chromatography (silica gel, 10 → 20% Et2O/hex) to yield (2»S',3i?)-l-azido-3-benzyloxy-4-penten-2-ol (3) (2.66 g, 66% yield) as a non-volatile clear liquid: R/= 4.8 (1 :4 EtOAc/hex); HRMS (ESI) (M+Na)+ calc. for C12H15N3O2 256.1056, obs. 256.1057; [α]D = -46.3° (c 1.50, CHCl3); 1H NMR (CDCl3, 300 MHz) δ 7.42-7.28 (m, 5H), 5.91-5.76 (m, IH), 5.46 (dd, J = 17.16, 1.42 Hz, IH), 5.42 (dd, J = 24.00, 1.37 Hz, IH), 4.65 (d, J = 11.67 Hz, IH), 4.39 (d, J = 11.67 Hz, IH), 3.88-3.80 (m, 2H), 3.44-3.40 (m, 2H), 2.22 (d, J = 3.60 Hz, IH); 13C NMR (CDCl3, 100 MHz) δ 137.88, 134.60, 128.66 (2C), 128.08 (2C), 128.05, 121.40, 81.39, 72.61, 70.70, 53.0; FTIR (NaCl): 3435, 2870, 2102, 1642, 1454, 1070 cm"1.
Figure imgf000095_0001
3 4
To a stirring solution of DAST (900 μL, 6.87 mmol) in benzene (3.2 mL) and pyridine (400 μL) in a plastic container at -10°C was added (2S,3.K)-l-azido-3- benzyloxy-4-penten-2-ol (3) (750 mg, 3.21 mmol) in small portions, and the reaction was stirred at this temperature for 48 hr followed by 6 hr at rt. The reaction mixture was slowly added to sat. aq. NaHCO3 (20 mL) at 0°C and was stirred for 10 min. The resulting aqueous mixture was extracted with DCM (3 x) and the combined organic layers were washed with 2 N HCl, dried over MgSO4, filtered and reduced under vacuum to yield a crude, which was purified by flash chromatography (silica gel, 1% Et2O/hex) to yield (3i?,4i?)-5-azido-4-fluoro-3-benzyloxy-pent-l-ene (4) (128 mg, 16.9% yield) as a nonvolatile clear liquid: R/= 0.63 (1 :9 EtOAC/Hex); [α]D = -11 -9° (c 1.50, CHCl3); 1H NMR (CDCl3, 400 MHz) δ 7.44-7.29 (m, 5H), 4.63 (dddd, J = 47.64, 7.07, 4.99, 3.32 Hz, IH), 5.49-5.42 (m, 2H), 4.70 (d, J = 11.95 Hz, IH), 4.57 (ddd, J = 7.07, 4.99, 3.32 Hz, IH), 4.44 (d, J = 11.90 Hz, IH), 4.03 (ddd, J = 16.87, 7.57, 5.04 Hz, IH), 3.64-3.52 (m, IH), 3.45 (ddd, J = 27.45, 13.63, 3.27 Hz, IH). 19F NMR (CDCl3, 282 MHz) -196.66 (dddd, J = 47.27, 27.08, 19.84, 16.89 Hz); 13C NMR (CDCl3, 100 MHz) δ 137.80, 133.09 (d, J = 5.30 Hz), 128.70 (2C), 128.09 (3C), 121.04, 93.33 (d, J = 181.54 Hz), 79.08 (d, J = 20.39 Hz), 70.92, 51.46 (d, J = 22.25 Hz). FTIR (NaCl): 2930, 2104, 1643, 1454, 1281, 1115, 1069 cm"1.
Figure imgf000095_0002
(3Λ,4i?)-5-azido-4-fluoro-3-benzyloxy-pent-l-ene (4) (128 mg, 0.543 mmol) was submitted to Procedure 2, followed by recrystallization from hot hexanes (2 x) to yield (2i?,3i?)-4-azido-2-benzyloxy-3-fluorobutanoic acid (5) (120 mg, 90%): [α]D = -56.9° (c 0.68, CHCl3); HRMS (ESI negative mode) (M-H) calc. for C11Hi2FN3O3 252.0790, obs. 252.0782; 1H NMR (CDCl3, 400 MHz), δ 10.55 (s, IH), 7.46-7.34 (m, 5H), 4.98 (dddd, J = 46.40, 7.57, 4.91, 2.92 Hz, IH), 4.94 (d, J = 11.47 Hz, IH), 4.55 (d, J = 11.51 Hz, IH), 4.17 (dd, J = 27.26, 2.86 Hz, IH), 3.77 (dt, J = 13.89, 13.66, 7.27 Hz, IH), 3.42 (ddd, J = 24.28, 13.20, 4.92 Hz, IH); 19F NMR (CDCl3, 376 MHz) δ-198.36 (dddd, J = 46.28, 27.22, 24.46, 14.15 Hz); 13C NMR (CDCl3, 100 MHz) δ 174.63 (d, J = 4.21 Hz), 136.37, 129.15 (2C), 129.07, 128.98 (2C), 91.53 (d, J = 182.59 Hz), 76.40 (d, J = 19.90 Hz), 73.96 (s), 50.87 (d, J = 25.13 Hz); FTIR (NaCl): 3151, 2098, 1753, 1407, 1283, 1112 cm"1.
Figure imgf000096_0001
3',4',3'",4'"-Tetra-dehydro-3,2',6',2'",6'"-penta-Cbz-neomycin (6, 50 mg, 0.41 mmol) and acid (5) were submitted to Procedure 3 to yield 3',4',3'",4'"-tetra- dehydro-l-(4-azido-3(JR)-fluoro-2(i?)-benzyloxy-butyryl)-3,2',6',2'",6'"-penta-Cbz- neomycin (7, 51.1 mg, 0.35 mmol, 85.6% yield): HRMS (ESI) (M+Na)+ calc. for C74H82FN9O21 1474.5502, obs. 1474.5450; FTIR (NaCl): 3332, 2919, 2477, 2105, 1695, 1531, 1454, 1245, 1114, 1048, 753, 698 cm"1.
Figure imgf000097_0001
3',4',3'",4'"-Tetra-dehydro-l-(4-azido-3(7?)-fluoro-2(i?)-benzyloxy- butyryl)-3,2',6',2'",6'"-penta-Cbz-neomycin (7, 51.1 mg, 0.35 mmol) was submitted to Procedure 4 to yield 3',4',3'",4'"-tetra-deoxy-l-(4-ammo-3(i?)-fluoro-2(i?)-hydroxy- butyryl)-neomycin acetate salt (8, 29.9 mg, 82.5% yield): HRMS (ESI) (M+H)+ calc. for C27H52FN7O11 670.3782, obs. 670.3773; [α]D = +33.9°(c 1.5, H2O); CLND 94.4% purity.
Example 25
Figure imgf000097_0002
OBn OBn
OBn
DAST "I) O3, DCM N3 ^~"^ ό i ' N3-
OH benzene, pyr F 2) NaH2PO4, NaCIO2 Ύ F Y OH
Figure imgf000097_0003
Starting from penta-l,4-dienol (5 g, 59.4 mmol) and using (S,S)-(+)- diisopropyl tartrate under the same reaction conditions as described in Example 24 the enantiomer ent-2 was obtained (4.9 g, 43% yield): [α]D = +35.7° (c 1.76, CHCl3). (2R, 3S)-l,2-Epoxy-3-benzyloxy-4-pentene (ent-2, 3.9g, 20.5 mmol) was submitted to the same reaction conditions described in Example 24 to yield the enantiomer (2R,3S)-\- azido-3-benzyloxy-4-penten-2-ol (ent-3, 2.75 g, 57% yield): [α]D = +47.3° (c 1.30, CHCl3). (2i?,35)-l-Azido-3-benzyloxy-4-penten-2-ol (ent-3) (500 mg, 2.14 mmol) was submitted to the same reactions as described in Example 24 to yield the enantiomer (3S,45)-5-azido-4-fluoro-3-benzyloxy-pent-l-ene (ent-4, 75.5 mg, 0.32 mmol, 15% yield, [α]o = +10.7°,c 1.50, CHCl3), which was submitted to the same reaction conditions as described in Example 24 to yield ent-5 (59 mg, 73% yield): [α]o= +58.6° (c 0.73, CHCl3).
Figure imgf000098_0001
3',4',3'",4'"-Tetra-dehydro-3,2',6',2'",6'"-penta-Cbz-neomycin (6, 50 mg, 0.41 mmol) and acid ent-5 were submitted to Procedure 3 to yield 3',4',3'",4'"- tetra-dehydro-l-(4-azido-3(5)-fluoro-2(4S)-benzyloxy-butyryl)-3,2',6',2'",6'"-penta- Cbz-neomycin (7, 54.2 mg, 0.37 mmol, 90% yield): HRMS (ESI) (M+Na)+ calc. for C74H82FN9O21 1474.5502, obs. 1474.5449; FTIR (NaCl): 3384, 2931, 2478, 2105, 1697, 1523, 1498, 1430, 1356, 1163, 1048, 752, 698 cm"1.
Figure imgf000098_0002
3',4',3'",4'"-Tetra-dehydro-l-(4-azido-3(5)-fluoro-2(5)-benzyloxy- butyryl)-3,2',6',2'",6'"-penta-Cbz-neomycin (7, 54.2 mg, 0.37 mmol) was submitted to Procedure 4 to yield 3',4',3"',4"'-tetra-deoxy-l-(4-amino-30S)-fluoro-2(S)-hydroxy- butyryl)-neomycin acetate salt (8, 15.7 mg, 71.4% yield): HRMS (ESI) (M+H)+ calc. for C27H52FN70, 1,670.3782, obs. 670.3780; [α]D = +41.9°(c 0.79, H2O); CLND 89.6% purity. Example 26
Figure imgf000099_0001
1 2 To a stirring solution of DMSO (690 μL, 9.65 mmol) in DCM (25 mL) at -78°C was added oxalyl chloride (3.21 mL of a 2.0 M solution in DCM, 6.43 mmol) and the reaction was stirred for 1 hr. A solution of (25,3i?)-l-azido-3-benzyloxy-4- penten-2-ol (1) (750 mg, 3.21 mmol) in DCM (1 mL) was added dropwise and the reaction mixture was stirred for 1 hr at -78°C. iV-Methyl morpholine (1.41 mL, 12.9 mmol) was added dropwise, and the reaction was stirred at -15°C for 2 hr. The reaction was quenched with phosphate buffer (0.1 M, pH 6.0) and the aqueous layer was separated. The organic layer was washed with the phosphate buffer (3 x), dried over Na2SO4, filtered and reduced under vacuum to give a brown residue. The residue was dissolved in Et2O, dried over MgSO4, filtered through a cotton plug, and reduced under vacuum to yield the crude ketone, which was dissolved in DCM (1 mL) and was added to a stirring solution of DAST (2 mL, 15.3 mmol) in DCM (3 mL) in a plastic vial at - 25°C. The reaction was allowed to slowly warm to rt and was stirred for 48 hr. The reaction mixture was then slowly poured into stirring sat. aq. NaHCO3 (20 mL) at 0°C, and was stirred for 10 min. The resulting aqueous mixture was extracted with DCM (3x), and the combined organic layers were dried over Na2SO4, filtered and reduced under vacuum to yield a crude, which was purified by flash chromatography (silica gel, 1% Et2θ/hex) followed by preparative TLC purification (silica gel, 0.5 mm, 5% Et2O/hex) to yield (Λ)-5-azido-4,4-difluoro-3-benzyloxy-pent-l-ene (2, 193 mg, 0.76 mmol, 24% yield), as a non-volatile clear liquid: R/= 0.72 (1 :4 EtOAc/hex); [α]o = - 23.8° (c 1.52, CHCl3); 1H NMR (CDCl3, 300 MHz) δ 7.44-7.31 (m, 5H), 5.89 (dddd, J = 16.88, 10.61, 7.11, 0.62 Hz, IH), 5.59-5.56 (m, IH), 5.53 (d, J = 10.74 Hz, IH), 4.71 (d, J = 11.67 Hz, IH), 4.50 (d, J = 11.66 Hz, IH), 4.14 (td, J = 14.25, 7.13, 7.13 Hz, IH), 3.64 (tq, J = 13.67, 13.67, 13.67, 11.19, 11.19 Hz, 2H); 19F NMR (CDCl3, 282 MHz) δ -116.63 (dtd, J = 257.62, 13.91, 13.90, 8.72 Hz), -111.27 (dtd, J = 257.59, 16.18, 16.16, 7.04 Hz); 13C NMR (CDCl3, 75 MHz) δ 137.14, 130.33 (t, J = 3.06, 3.06 Hz), 128.71 (2C), 128.27, 128.20 (2C), 122.78, 120.69 (dd, J = 249.89, 246.83 Hz), 78.87 (dd, J = 30.35, 25.35 Hz), 71.48 (d, J = 0.48 Hz), 51.47 (dd, J = 30.26, 25.92 Hz); FTIR (NaCl): 2928, 2108, 1455, 1292, 1091 cm"1.
Figure imgf000100_0001
(i?)-5-Azido-4,4-difmoro-3-benzyloxy-pent-l-ene (2, 193 mg, 0.76 mmol) was submitted to Procedure 2, followed by washing with cold hexanes (3x) at - 20°C to yield (3) (139 mg, 67.6% yield): [α]D = -32.4° (c 0.80, CHCl3); HRMS (ESI negative mode) (M-H) for CnHnF2N3O3 270.0696, obs. 270.06924; 1H NMR (CDCl3, 400 MHz) δ 7.46-7.32 (m, 5H), 6.48 (s, IH), 4.84 (d, J = 11.30 Hz, IH), 4.67 (d, J = 11.30 Hz, IH), 4.37 (dd, J = 12.23, 9.78 Hz, IH), 3.75 (dd, J = 14.67, 12.35 Hz, 2H); 19F NMR (CDCl3, 376 MHz) δ -112.61 (qd, J = 260.95, 12.30, 12.29, 12.29 Hz), - 109.68 (dtd, J = 260.79, 14.75, 14.68, 9.94 Hz); 13C NMR (CDCl3, 100 MHz) δ 170.84, 135.48, 129.01, 128.94 (2C), 128.78 (2C), 1 19.59 (t, J = 251.58, 251.58 Hz), 76.56 (dd, J = 29.86, 27.24 Hz), 74.34, 51.58 (dd, J = 28.94, 26.76 Hz). FTIR (NaCl): 3337, 2929, 21 12, 1738, 1455, 1292, 1210, 1119 cm"1.
Figure imgf000100_0002
3',4',3'",4'"-Tetra-dehydro-3,2',6',2'",6'"-penta-Cbz-neomycin (4, 50 mg, 0.41 mmol) and acid (3) were submitted to Procedure 3 to yield 3',4',3'",4'"-tetra- dehydro- 1 -(4-azido-3 ,3 -bis-fluoro-2(7?)-benzyloxy-butyryl)-3 ,2 ' ,6 ' ,2 '",6 ' "-penta-Cbz- neomycin (5, 45.1 mg, 0.31 mmol, 75% yield): HRMS (ESI) (M+Na)+ calc. for C74H81F2N9O2I 1492.5407, obs. 1492.5343; FTIR (NaCl): 3412, 2938, 2476, 2110, 1693, 1497, 1428, 1428, 1356, 1163, 1111, 1047, 753, 697 cm"1.
Figure imgf000101_0001
3',4',3'",4'"-Tetra-dehydro-l-(4-azido-3,3-bisfluoro-2(i?)-benzyloxy- butyryl)-3,2',6',2'",6'"-penta-Cbz-neomycin (5, 45.1 mg, 0.31 mmol) was submitted to Procedure 4 to yield 3',4',3'",4'"-tetra-deoxy-l-(4-amino-3,3-bisfluoro-2(i?)-hydroxy- butyryl)-neomycin (6, 28.1 mg, 87.5% yield): HRMS (ESI) (M+H)+ calc. for C27H51F2N7O,,688.3688, obs. 688.3687; [α]D = +38.0° (c 1.41, H2O); CLND 97.5% purity.
Example 27
Figure imgf000101_0002
ent-1 ent-2 ^V^ ent-3 (2i?,35)-l-Azido-3-benzyloxy-4-penten-2-ol (ent-1, 500 mg, 2.14 mmol) was submitted to the same reaction conditions described in Example 26 to yield (,S)-5-azido-4,4-difluoro-3-benzyloxy-pent-l-ene (ent-2, 1 14 mg, 21% yield, [α]o = +27.9° (c 3.14, CHCl3)). Ent-2 (75.5 mg, 0.32 mmol) was submitted to Procedure 2 to yield (5)-4-azido-2-benzyloxy-3,3-difluorobutanoic acid (ent-3, 34.8 mg, 43% yield, [α]D = +36.4° (c 0.80, CHCl3).
Figure imgf000102_0001
3',4',3'",4'"-Tetra-dehydro-3,2',6',2'",6'"-penta-Cbz-neomycin (4, 50 mg, 0.41 mmol) and acid ent-3 were submitted to Procedure 3 to yield 3',4',3'",4'"- tetra-dehydro-l-(4-azido-3,3-bisfluoro-2(lS)-benzyloxy-butyryl)-3,2',6',2'",6'"-penta- Cbz-neomycin (5, 31.2 mg, 0.20 mmol, 52% yield): HRMS (ESI) (M+Na)+ calc. for
C74H8]F2N9O2I, 1492.5407, obs. 1492.5348; FTIR (NaCl): 3417, 2932, 2477, 2110,
2694, 1497, 1428, 1357, 1164, 1107, 1048, 753, 698 cm -1
Figure imgf000102_0002
3',4',3"',4'"-Tetra-dehydro-l-(4-azido-3,3-bisfluoro-2(5)-benzyloxy- butyryl)-3,2',6',2'",6'"-penta-Cbz-neomycin (5, 31.2 mg, 0.20 mmol) was submitted to Procedure 4 to yield 3',4',3'",4'"-tetra-deoxy-l-(4-amino-3,3-bisfluoro-2(5)-hydroxy- butyryl)-neomycin acetate salt (6, 20.2 mg, 52% yield): HRMS (ESI) (M+Na)+ calc. for C27H5]F2N7On, 688.36796, obs. 688.36874; [α]D = +41.0° (c 1.01, H2O); CLND 97.9% purity.
Example 28
Figure imgf000103_0001
1 2
To a stirring solution of (25,3i?)-l-azido-3-benzyloxy-4-penten-2-ol (1) (250 μL, 1.07 mmol) in THF (50 mL) under argon was added tetrabutylammonium iodide (42 mg, 0.11 mmol) followed by benzyl bromide (155 μL, 1.27 mmol) and the reaction was cooled to 0°C. Sodium hydride (60% in mineral oil, 47 mg, 1.18 mmol) was added in small portions and the reaction was stirred overnight with warming to rt. The reaction was quenched with MeOH, filtered through Celite, and washed with Et2O. The organic solvent was removed under vacuum to give an oily residue, which was purified by flash chromatography (silica gel, 2% Et2O/hex) to yield (3i?,45)-5-azido- 3,4-bisbenzyloxy-pent-l-ene (2, 237 mg, 65% yield) as a clear non-volatile liquid: Rf=
0.62 (1 :4 EtOAc/hex); [α]D = -6.1 ° (c 1.50, CHCl3); 1H NMR (CDCl3, 300 MHz) δ 7.35-7.24 (m, 10H), 5.81 (ddd, J = 17.15, 10.58, 7.45 Hz, IH), 5.37 (ddd, J = 5.70,
1.65, 0.86 Hz, IH), 5.33 (ddd, J = 12.07, 1.44, 0.81 Hz, IH), 4.63 (s, 2H), 4.61 (d, J =
11.87 Hz, IH), 4.35 (d, J = 11.78 Hz, IH), 3.90 (tdd, J = 7.37, 5.65, 0.79, 0.79 Hz, IH), 3.60 (ddd, J = 6.39, 5.69, 3.64 Hz, IH), 3.43 (dd, J = 12.93, 6.42 Hz, IH), 3.35 (dd, J = 12.93, 3.60 Hz, IH); 13C NMR (CDCl3, 75 MHz) δ 138.25, 138.01, 135.43, 128.60 (4C), 128.29 (2C), 128.02, 127.99 (2C), 127.87, 119.97, 80.76, 80.23, 73.33, 70.79, 51.69; FTIR (NaCl): 2867, 2100, 1606, 1454, 1286, 1095, 1073.
Figure imgf000103_0002
3
(37?,45)-5-azido-3,4-bis-benzyloxy-pent-l-ene (2, 237 mg, 0.69 mmol) was submitted to Procedure 2 to yield (25',35)-4-azido-2,3-bis-benzyloxybutanoic acid (3, 187.7 mg, 75% yield): [α]D = -15.1 ° (c 1.05, CHCl3); HRMS (ESI negative mode) (M-H) calc. for Ci8H19N3O4 340.1303, obs. 340.1296; 1H NMR (CDCl3, 300 MHz) δ 7.24 (s, IH), 7.38-7.33 (m, 10H), 4.79 (d, J = 11.61 Hz, IH), 4.66 (s, 2H), 4.56 (d, J = 11.61 Hz, IH), 4.20 (d, J = 4.24 Hz, IH), 3.98 (td, J = 6.56, 4.30, 4.30 Hz, IH), 3.58 (dd, J = 13.04, 6.62 Hz, IH), 3.42 (dd, J = 13.04, 4.31 Hz, IH); 13C NMR (CDCl3, 75 MHz) δ 175.57, 137.92, 137.34, 129.44 (2C), 129.36 (2C), 129.15, 129.04 (2C), 128.98 (2C), 128.94, 79.71, 77.651, 74.04, 73.89, 51.65; FTIR (NaCl): 3000, 2918, 2103, 1722, 1455, 1284, 1110 cm-1.
Figure imgf000104_0001
3',4',3'",4'"-Tetra-dehydro-3,2',6',2'",6'"-penta-Cbz-neomycin (4, 50 mg, 0.41 mmol) and acid 3 were submitted to Procedure 3 to yield 3',4',3'",4'"-tetra- dehydro- 1 -(4-azido-3 ,2-(5,5)-bis-benzyloxy-butyryl)-3 ,2 ' ,6' ,2 '",6 ' "-penta-Cbz- neomycin (5, 37.9 mg, 0.25 mmol, 60% yield): HRMS (ESI) (M+Na)+ calc. for C8,H89N9O22l562.6014, obs. 1562.5939.
Figure imgf000104_0002
3',4',3'",4'"-Tetra-dehydro-l-(4-azido-3,2-(,S,5)-bis-benzyloxy-butyryl)- 3,2',6',2'",6'"-penta-Cbz-neomycin (5, 37.9 mg, 0.25 mmol) was submitted to Procedure 4 to yield 3',4',3'",4'"-tetra-deoxy-l-(4-amino-3,2-(5',5)-bis-hydroxy- butyryl)-neomycin acetate salt (6, 14.3 mg, 56.5% yield): HRMS (ESI) (M+Na)+ calc. for C27H53N7O]2, 690.3644, obs. 690.3654; [α]D = +32.9° (c 0.72, H2O); CLND 96.5% purity.
Example 29 OBn OBn OBn
Λ BnBr Λ I) O3, DCM .0
OH Bu4NI OBn 2) NaH2PO4, NaCIO2 OBn OH
Figure imgf000105_0001
(2Λ,3S)-l-azido-3-benzyloxy-4-penten-2-ol (ent-1, 250 mg, 1.07 mmol) was submitted to the same reaction conditions as described in Example 28 to yield (3S,4ft)-5-azido -3,4-bis-benzyloxy-pent-l-ene (ent-2, 322mg, 59% yield): [α]D= +7.9°
(c 1.50, CHCl3). Ent-2 (178 mg, 0.55 mmol) was submitted to Procedure 2 to yield ent-3 (144 mg, 77% yield): [α]D= +15.2° (c 0.81, CHCl3).
ΛNHCbz
Figure imgf000105_0003
Figure imgf000105_0002
3',4',3'",4'"-Tetra-dehydro-3,2',6',2'",6'"-penta-Cbz-neomycin (4, 50 mg, 0.41 mmol) and acid ent-3 were submitted to Procedure 3 to yield 3',4',3'",4'"- tetra-dehydro-l-(4-azido-3,2-(i?,i?)-bis-benzyloxy-butyryl)-3,2',6',2'",6'"-penta-Cbz- neomycin (5, 37.9 mg, 0.25 mmol, 60% yield). HRMS (ESI) (M+Na)+ calc. for C8IH89N9O22, 1562.6014. obs. 1562.5947.
Figure imgf000105_0004
3',4',3'",4'"-Tetra-dehydro-l-(4-azido-3,2-(i?,i?)-bis-benzyloxy- butyryl)-3,2',6',2'",6'"-penta-Cbz-neomycin (5, 37.9 mg, 0.25 mmol) was submitted to Procedure 4 to yield 3',4',3'",4'"-tetra-deoxy-l-(4-amino-3,2-(i?,i?)-bis-hydroxy- butyryl)-neomycin acetate salt (6, 15.4 mg, 61% yield): HRMS (ESI) (M+Na)+ calc. for C27H53N7Oi2, 690.3644, obs. 690.3641; [α]D = +44.0° (c 0.77, H2O); CLND 94.7% purity.
MIC Assay Protocol
Minimum inhibitory concentrations (MIC) were determined by reference Clinical and Laboratory Standards Institute (CLSI) broth microdilution methods per M7-A7 [2006]. Quality control ranges utilizing E. coli ATCC 25922, P. aeruginosa ATCC 27853 and S. aureus ATCC 29213, and interpretive criteria for comparator agents were as published in CLSI Ml 00-Sl 7 [2007]. Briefly, serial two-fold dilutions of the test compounds were prepared at 2X concentration in Mueller Hinton Broth. The compound dilutions were mixed in 96-well assay plates in a 1 :1 ratio with bacterial inoculum. The inoculum was prepared by suspension of a colony from an agar plate that was prepared the previous day. Bacteria were suspended in sterile saline and added to each assay plate to obtain a final concentration of 5x1 O^ CFU/mL. The plates were incubated at 35C for 20 hours in ambient air. The MIC was determined to be the lowest concentration of the test compound that resulted in no visible bacterial growth as compared to untreated control. Data for certain representative compounds is shown in Table 1 below.
Table 1
Figure imgf000106_0001
Figure imgf000107_0001
* AECOOOl is ATCC25922 and APAEOOl is ATCC27853. ** MIC Key:
MICs of 1.0 μg/mL or less = A MICs of greater than 1.0 μg/mL to 16.0 μg/mL = B MICs of greater than 16.0 μg/mL = C
All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification are incorporated herein by reference, in their entirety to the extent not inconsistent with the present description.
From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. Accordingly, the invention is not limited except as by the appended claims.

Claims

CLAIMS"What is claimed is:
1. A compound having the following structure (I):
Figure imgf000109_0001
(I) or a stereoisomer, prodrug or pharmaceutically acceptable salt thereof, wherein:
Qi is hydroxyl, a protected hydroxyl, amino or a protected amino group; Q2 is hydrogen, optionally substituted alkyl,
Figure imgf000109_0002
Figure imgf000109_0003
Figure imgf000110_0001
Figure imgf000110_0002
Figure imgf000110_0003
Figure imgf000110_0004
Figure imgf000110_0005
Figure imgf000111_0001
each Ri and R2 is, independently, hydrogen or an amino protecting group; each R3 is, independently, hydrogen or a hydroxyl protecting group; each R4, R5, R7 and Rg is, independently, hydrogen or CpC6 alkyl optionally substituted with one or more halogen, hydroxyl or amino; each R6 is, independently, hydrogen, halogen, hydroxyl, amino or Cj-C6 alkyl; or R4 and R5 together with the atoms to which they are attached can form a heterocyclic ring having from 4 to 6 ring atoms, or R5 and one R6 together with the atoms to which they are attached can form a heterocyclic ring having from 4 to 6 ring atoms, or R4 and one R6 together with the atoms to which they are attached can form a carbocyclic ring having from 3 to 6 ring atoms, or R7 and R8 together with the atom to which they are attached can form a heterocyclic ring having from 4 to 6 ring atoms; each R9 is, independently, hydrogen, hydroxyl, amino or Ci-C6 alkyl; each Rjo is, independently, hydrogen, halogen, hydroxyl, amino or Ci-C6 alkyl; or R9 and one Ri0 together with the atoms to which they are attached can form a heterocyclic ring having from 4 to 6 ring atoms; n is an integer from 0 to 4;
Zi and Z2 are both hydrogen or Zi and Z2 form a double bond; and Z3 and Z4 are both hydrogen or Z3 and Z4 form a double bond.
2. A compound of claim 1 wherein each R1, R2 and R3 are hydrogen.
3. A compound of claim 1 or 2 wherein Qi is amino.
4. A compound of claim 1 or 2 wherein Qi is hydroxyl.
5. A compound of any one of claims 1-4 wherein Zi and Z2 form a double bond.
6. A compound of any one of claims 1-4 wherein Zi and Z2 are both hydrogen.
7. A compound of any one of claims 1-6 wherein Z3 and Z4 form a double bond.
8. A compound of any one of claims 1-6 wherein Z3 and Z4 are both hydrogen.
9. A compound of any one of claims 1-8 wherein Q2 is:
5
Figure imgf000112_0001
wherein:
R4 is hydrogen; and R5 is hydrogen.
10. A compound of claim 9 wherein each R6 is hydrogen.
11. A compound of claim 10 wherein Q2 is:
Figure imgf000113_0001
12. A compound of claim 9 wherein at least one R6 is halogen.
13. A compound of any one of claims 1-8 wherein Q2 is:
Figure imgf000113_0002
wherein:
R4 is hydrogen; and
R5 and one R6 together with the atoms to which they are attached form a heterocyclic ring having from 4 to 6 ring atoms.
14. A compound of claim 13 wherein Q2 is:
Figure imgf000114_0001
15. A compound of claim 13 wherein at least one R6 is halogen.
16. A compound of any one of claims 1-8 wherein Q2 is:
Figure imgf000114_0002
wherein R4 and R5 together with the atoms to which they are attached form a heterocyclic ring having from 4 to 6 ring atoms.
17. A compound of claim 16 wherein each R6 is hydrogen.
18. A compound of claim 17 wherein Q2 is:
Figure imgf000115_0001
Figure imgf000115_0002
19. A compound of claim 16 wherein at least one R6 is halogen.
20. A compound of any one of claims 1-8 wherein Q2 is:
Figure imgf000115_0003
wherein:
R.5 is hydrogen; and
R4 and one R6 together with the atoms to which they are attached form a carbocyclic ring having from 3 to 6 ring atoms.
21. A compound of claim 20 wherein Q2 is:
Figure imgf000116_0001
22. A compound of claim 20 wherein at least one R6 is halogen.
23. A compound of any one of claims 1-8 wherein Q2 is:
Figure imgf000116_0002
wherein:
R4 is hydrogen; R7 is hydrogen; and Rs is hydrogen.
24. A compound of claim 23 wherein each R6 is hydrogen;
25. A compound of claim 24 wherein Q2 is:
Figure imgf000117_0001
26. A compound of claim 23 wherein at least one R6 is halogen.
27. A compound of any one of claims 1-8 wherein Q2 is:
Figure imgf000117_0002
wherein:
R4 and one R6 together with the atoms to which they are attached form a carbocyclic ring having from 3 to 6 ring atoms; R7 is hydrogen; and Rg is hydrogen.
28. A compound of claim 27 wherein Q2 is:
Figure imgf000118_0001
Figure imgf000118_0002
Figure imgf000118_0004
Figure imgf000118_0003
Figure imgf000118_0005
29. A compound of claim 27 wherein at least one R6 is halogen.
30. A compound of any one of claims 1-8 wherein Q2 is:
Figure imgf000118_0006
wherein R5 is hydrogen.
31. A compound of claim 30 wherein each R6 is hydrogen.
32. A compound of claim 31 wherein Q2 is:
Figure imgf000119_0001
33. A compound of claim 30 wherein at least one R6 is halogen.
34. A compound of any one of claims 1-8 wherein Q2 is:
Figure imgf000119_0002
wherein:
R7 is hydrogen; and R8 is hydrogen.
35. A compound of claim 34 wherein each R6 is hydrogen.
36. A compound of claim 35 wherein Q2 is:
Figure imgf000120_0001
37. A compound of claim 34 wherein at least one R6 is halogen.
38. A compound of any one of claims 1-8 wherein Q2 is:
Figure imgf000120_0002
wherein R5 is hydrogen.
39. A compound of claim 38 wherein each R6 is hydrogen.
40. A compound of claim 38 wherein at least one R6 is halogen.
41. A compound of any one of claims 1 -8 wherein Q2 is:
Figure imgf000120_0003
wherein:
R7 is hydrogen; and Rg is hydrogen.
42. A compound of claim 41 wherein each R6 is hydrogen,
43. A compound of claim 41 wherein at least one R6 is halogen.
44. A compound of any one of claims 1 -8 wherein Q2 is:
Figure imgf000121_0001
wherein R5 is hydrogen.
45. A compound of claim 44 wherein each R6 is hydrogen.
46. A compound of claim 44 wherein at least one R6 is halogen.
47. A compound of any one of claims 1-8 wherein Q2 is:
Figure imgf000121_0002
wherein R9 is hydrogen.
48. A compound of claim 47 wherein each R]0 is hydrogen.
49. A compound of claim 47 wherein at least one Ri0 is halogen.
50. A compound of any one of claims 1 -8 wherein Q2 is:
Figure imgf000122_0001
wherein:
R7 is hydrogen; and R8 is hydrogen.
51. A compound of claim 50 wherein each Rio is hydrogen.
52. A compound of claim 50 wherein at least one Rio is halogen.
53. A compound of any one of claims 1 -8 wherein Q2 is:
Figure imgf000122_0002
wherein R4 is hydrogen.
54. A compound of claim 53 wherein each R6 is hydrogen.
55. A compound of claim 53 wherein at least one R6 is halogen.
56. A compound of claim 53 wherein Q2 is -C(=O)H.
57. A compound of any one of claims 1-8 wherein Q2 is hydrogen.
58. A compound of any one of claims 1-8 wherein Q2 is optionally substituted alkyl.
59. A compound of claim 58 wherein Q2 is unsubstituted.
60. A compound of claim 58 wherein Q2 is substituted with one or more halogen, hydroxyl or amino.
61. A compound of any one of claims 1 -60 having the configuration:
Figure imgf000123_0001
62. A pharmaceutical composition comprising a compound of any one of claims 1-61, or a stereoisomer, pharmaceutically acceptable salt or prodrug thereof, and a pharmaceutically acceptable carrier, diluent or excipient.
63. A method of treating a bacterial infection in a mammal comprising administering to the mammal an effective amount of a compound of any one of claims 1-61 or a pharmaceutical composition of claim 62.
PCT/US2009/056407 2008-09-10 2009-09-09 Antibacterial aminoglycoside analogs WO2010030704A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/044,234 US8399419B2 (en) 2008-09-10 2011-03-09 Antibacterial aminoglycoside analogs

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US9567308P 2008-09-10 2008-09-10
US61/095,673 2008-09-10

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/044,234 Continuation US8399419B2 (en) 2008-09-10 2011-03-09 Antibacterial aminoglycoside analogs

Publications (2)

Publication Number Publication Date
WO2010030704A2 true WO2010030704A2 (en) 2010-03-18
WO2010030704A3 WO2010030704A3 (en) 2010-05-20

Family

ID=41723042

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/056407 WO2010030704A2 (en) 2008-09-10 2009-09-09 Antibacterial aminoglycoside analogs

Country Status (2)

Country Link
US (1) US8399419B2 (en)
WO (1) WO2010030704A2 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7893039B2 (en) 2005-12-02 2011-02-22 Isis Pharmaceuticals, Inc. Antibacterial 4,5-substituted aminoglycoside analogs having multiple substituents
US8318685B2 (en) 2010-11-17 2012-11-27 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8367625B2 (en) 2008-10-09 2013-02-05 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8372813B2 (en) 2008-10-09 2013-02-12 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8377896B2 (en) 2008-09-10 2013-02-19 Isis Pharmaceuticals, Inc Antibacterial 4,6-substituted 6′, 6″ and 1 modified aminoglycoside analogs
US8383596B2 (en) 2007-11-21 2013-02-26 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8399419B2 (en) 2008-09-10 2013-03-19 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8481502B2 (en) 2009-10-09 2013-07-09 Achaogen, Inc. Antibacterial aminoglycoside analogs
CN103204887A (en) * 2013-04-11 2013-07-17 北京化工大学 Method for synthesizing dibekacin and arbekacin
US8492354B2 (en) 2009-05-15 2013-07-23 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8524689B2 (en) 2009-05-15 2013-09-03 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8524675B2 (en) 2009-05-15 2013-09-03 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8653042B2 (en) 2009-05-15 2014-02-18 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8658606B2 (en) 2009-05-15 2014-02-25 Achaogen, Inc. Antibacterial aminoglycoside analogs

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2148884A1 (en) * 2007-04-10 2010-02-03 Achaogen, Inc. Antibacterial 1,4,5-substituted aminoglycoside analogs
US11407721B2 (en) 2013-02-19 2022-08-09 Amgen Inc. CIS-morpholinone and other compounds as MDM2 inhibitors for the treatment of cancer
JOP20200296A1 (en) * 2013-06-10 2017-06-16 Amgen Inc Processes of Making and Crystalline Forms of a MDM2 Inhibitor

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3808198A (en) * 1972-09-07 1974-04-30 Bristol Myers Co Lividomycin b derivatives
US3860574A (en) * 1972-12-06 1975-01-14 Bristol Myers Co Derivatives of neomycin b and neomycin c

Family Cites Families (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR1361393A (en) 1963-03-29 1964-05-22 Rit Rech Ind Therapeut Basic antibiotic derivatives and their preparation process
US3897412A (en) 1972-05-04 1975-07-29 Bristol Myers Co Paromomycin antibiotic derivatives
JPS5632999B2 (en) 1972-05-04 1981-07-31
US3896106A (en) 1972-06-26 1975-07-22 Bristol Myers Co Antibiotic derivatives
FR2209546B1 (en) 1972-12-06 1977-09-02 Bristol Myers Co
JPS5652035B2 (en) 1973-02-06 1981-12-09
JPS553358B2 (en) 1973-03-13 1980-01-24
US4170642A (en) 1973-10-01 1979-10-09 Zaidan Hojin Biseibutsu Kagaku Kenkyu Kai Derivatives of kanamycin A
US4021601A (en) 1974-04-11 1977-05-03 Societa' Farmaceutici Italia S.P.A. Paromomycin derivatives and process for the preparation thereof
DE2515630C2 (en) 1974-05-28 1982-11-18 Società Farmaceutici Italia S.p.A., 20146 Milano 4-0- (6-Amino-6-deoxy-alpha-D-glucopyranosyl) -5-0 [3-0 (2,6-diamino-2,6-dideoxy-β-L-idophyranosyl) -β-D -ribofuranosyl] -2-deoxy-streptamine, process for its preparation and pharmaceuticals containing this compound
JPS5850235B2 (en) 1974-12-11 1983-11-09 明治製菓株式会社 Method for producing 3',4'-α-epoxy-ribostamycin or -kanamycin B
JPS6055520B2 (en) 1976-02-17 1985-12-05 興和株式会社 Method for producing lividomycin B derivatives
US4066753A (en) 1976-05-27 1978-01-03 Canadian Patents And Development Limited Neomycin and paromomycin derivatives
US4424343A (en) 1977-04-28 1984-01-03 Bristol Myers Company Preparation of 1-N- ω-amino-α-hydroxyalkanoyl!kanamycin polysilylates and products
US4347354A (en) 1977-04-28 1982-08-31 Bristol-Myers Company Preparation of 1-N-[ω-amino-α-hydroxyalkanoyl]aminoglycoside polysilylated antibiotics and products obtained therefrom
CA1271744A (en) 1978-02-21 1990-07-17 Stephen Hanessian Preparation of aminoglycosides
GB1600457A (en) 1978-04-26 1981-10-14 Bristol Myers Co Process for the preparation of 1-n-acylaminoglycosides
JPS5515445A (en) 1978-07-21 1980-02-02 Kyowa Hakko Kogyo Co Ltd Introduction of amino-protecting group by microorganism
DE2936120A1 (en) 1978-09-14 1980-03-27 Sandoz Ag NEW AMINOGLYCOSIDES, THEIR PRODUCTION AND THEIR USE AS AN ANTIMICROBIAL AGENTS
ZA803031B (en) 1979-06-07 1981-05-27 Erba Farmitalia Paromomycin derivatives
US4247687A (en) 1979-06-12 1981-01-27 Farmitalia Carlo Erba S.P.A. Aminoglycoside antibiotic derivatives and process for their preparation
BE886393A (en) 1980-01-30 1981-03-16 Erba Farmitalia PAROMOMYCIN DERIVATIVES
GB2068366B (en) 1980-01-30 1983-06-02 Erba Farmitalia Paromycin derivative
JPS57501084A (en) 1980-07-28 1982-06-24
DE3206725A1 (en) 1981-05-13 1982-12-02 Merck Patent Gmbh, 6100 Darmstadt PERSONALLY SOLUBLE SALTS OF AMINOGLYCOSIDANTIBIOTICS
DE3405326A1 (en) 1984-02-15 1985-08-22 Sandoz-Patent-GmbH, 7850 Lörrach New aminoglycosides, process for the preparation thereof and the use thereof
DE3431534A1 (en) 1984-08-28 1986-03-13 Merck Patent Gmbh, 6100 Darmstadt FLAVANONE DERIVATIVES
IT1225484B (en) 1987-11-27 1990-11-14 Pierrel Spa SYNTHESIS PROCEDURE OF AMIKACINA
US5470836A (en) 1988-04-29 1995-11-28 Farmitalia Carlo Erba S.R.L. Paromomycin or its derivatives or salts thereof in combination with sodium stirogluconate for parenteral treatment of human parasitic diseases
CA2085367A1 (en) 1990-07-30 1992-01-31 William G. Tarpley Hiv-transactivators to treat aids
US5534408A (en) 1993-09-24 1996-07-09 University Of Massachusetts Medical Center 2-deoxystreptamine aminoglycoside inhibition of HIV RRE/Rev binding
WO1994009792A1 (en) 1992-10-23 1994-05-11 University Of Massachusetts Medical Center Small molecule inhibition of rna/ligand binding
EP0865283A4 (en) 1995-09-08 2005-07-20 Anadys Pharmaceuticals Inc 2-deoxystreptamine as a molecular scaffold for preparing functionally and spacially diverse molecules and pharmaceutical compositions thereof
IL127773A0 (en) 1998-12-28 1999-10-28 Yeda Res & Dev Saccharide conjugates and pharmaceutical compositions comprising them
US6541456B1 (en) * 1999-12-01 2003-04-01 Isis Pharmaceuticals, Inc. Antimicrobial 2-deoxystreptamine compounds
IT1317735B1 (en) 2000-01-26 2003-07-15 Nicox Sa SALTS OF ANTIMICROBIAL AGENTS.
EP1219306A1 (en) 2000-12-29 2002-07-03 Nicox S.A. Compositions comprising cyclodextrins and NO- releasing drugs
US20050085432A1 (en) 2001-12-26 2005-04-21 Aviva Lapidot Methods of using conjugates of saccharides and acetamidino or guanidino compounds for treating bacterial infections
WO2003101405A2 (en) 2002-03-14 2003-12-11 President And Fellows Of Harvard College Aminoglycoside antibiotics and methods of using same
US7037936B2 (en) 2002-06-17 2006-05-02 Signal Pharmaceuticals, Llc. Compounds useful for the treatment of cancer, compositions thereof and methods therewith
US7160992B2 (en) 2003-04-28 2007-01-09 Yeda Research And Development Co., Ltd. Amino-modified polysaccharides and methods of generating and using same
US7635685B2 (en) 2003-07-03 2009-12-22 Technion Research & Development Foundation Ltd. Bifunctional antibiotics for targeting rRNA and resistance-causing enzymes and for inhibition of anthrax lethal factor
US20050004052A1 (en) 2003-07-03 2005-01-06 Technion Research & Development Foundation Ltd. Bifunctional antibiotics for targeting rRNA and resistance-causing enzymes
WO2005041984A1 (en) 2003-10-31 2005-05-12 Steele Philip M Compositions and treatments for envelope virus infections
WO2006052930A1 (en) 2004-11-05 2006-05-18 Isis Pharmaceuticals, Inc. Antimicrobial 2-deoxystreptamine compounds
WO2007028012A2 (en) 2005-09-01 2007-03-08 Isis Pharmaceuticals, Inc. Antibacterial 6'-n-modified 4,5-substituted aminoglycoside analogs
CA2632968A1 (en) 2005-12-02 2007-06-07 Isis Pharmaceuticals, Inc. Antibacterial 4,5-substituted aminoglycoside analogs having multiple substituents
UY30118A1 (en) 2006-01-31 2007-06-29 Tanabe Seiyaku Co AMIS TRISUSTITUDE COMPUTER
CA2657227A1 (en) 2006-07-14 2008-01-17 Novartis Ag Pyrimidine derivatives as alk-5 inhibitors
EP1953171A1 (en) 2007-02-02 2008-08-06 Eidgenössische Technische Hochschule Zürich Aminoglycoside antibiotics targeting bacterial 16S ribosomal RNA
EP2148884A1 (en) 2007-04-10 2010-02-03 Achaogen, Inc. Antibacterial 1,4,5-substituted aminoglycoside analogs
JP4992044B2 (en) 2007-08-30 2012-08-08 Jfeスチール株式会社 Chromate-free coated hot-dip galvanized steel sheet and method for producing the same
DK2217610T3 (en) 2007-11-21 2017-02-13 Achaogen Inc ANTIBACTERIAL AMINOGLYCOSIDE ANALOGICAL COMPOUNDS
WO2010030690A1 (en) 2008-09-10 2010-03-18 Isis Pharmaceuticals, Inc. Antibacterial 4,6-substituted 6', 6" and 1 modified aminoglycoside analogs
WO2010030704A2 (en) 2008-09-10 2010-03-18 Achaogen, Inc. Antibacterial aminoglycoside analogs
WO2010042850A1 (en) 2008-10-09 2010-04-15 Achaogen, Inc. Antibacterial aminoglycoside analogs
WO2010042851A1 (en) 2008-10-09 2010-04-15 Achaogen, Inc. Antibacterial aminoglycoside analogs
CA2761756A1 (en) 2009-05-14 2010-11-18 Achaogen, Inc. Treatment of urinary tract infections with antibacterial aminoglycoside compounds
AR076584A1 (en) 2009-05-14 2011-06-22 Achaogen Inc METHOD TO TREAT BACTERIAL INFECTION. AMINOGLYCOSID DOSAGE REGIMES. AN AMINOGLUCOSIDE
CA2948868A1 (en) 2009-05-14 2010-11-18 Achaogen, Inc. Treatment of klebsiella pneumoniae infections with antibacterial aminoglycoside compounds
WO2010132768A1 (en) 2009-05-15 2010-11-18 Achaogen, Inc. Antibacterial derivatives of sisomicin
WO2010132765A2 (en) 2009-05-15 2010-11-18 Achaogen, Inc. Antibacterial aminoglycoside analogs
WO2010132760A1 (en) 2009-05-15 2010-11-18 Achaogen, Inc. Antibacterial derivatives of tobramycin
WO2010132759A1 (en) 2009-05-15 2010-11-18 Achaogen, Inc. Antibacterial derivatives of dibekacin
WO2010132757A2 (en) 2009-05-15 2010-11-18 Achaogen, Inc. Antibacterial aminoglycoside analogs
WO2010147836A1 (en) 2009-06-17 2010-12-23 Achaogen, Inc. Combination therapies using antibacterial aminoglycoside compounds
WO2011044502A1 (en) 2009-10-09 2011-04-14 Achaogen, Inc. Antibacterial aminoglycoside analogs
WO2011044503A1 (en) 2009-10-09 2011-04-14 Achaogen, Inc. Antibacterial aminoglycoside analogs
WO2011044498A1 (en) 2009-10-09 2011-04-14 Achaogen, Inc. Antibacterial aminoglycoside analogs
JP2013507391A (en) 2009-10-09 2013-03-04 アカオジェン インコーポレイテッド Antiviral aminoglycoside analogues
WO2011044538A1 (en) 2009-10-09 2011-04-14 Achaogen, Inc. Antibacterial aminoglycoside analogs
AR083879A1 (en) 2010-11-17 2013-03-27 Achaogen Inc ANTIBACTERIAL AMINOGLYCOSID ANALOGS, METHODS OF PREPARATION AND USE AS THERAPEUTIC AGENTS

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3808198A (en) * 1972-09-07 1974-04-30 Bristol Myers Co Lividomycin b derivatives
US3860574A (en) * 1972-12-06 1975-01-14 Bristol Myers Co Derivatives of neomycin b and neomycin c

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7893039B2 (en) 2005-12-02 2011-02-22 Isis Pharmaceuticals, Inc. Antibacterial 4,5-substituted aminoglycoside analogs having multiple substituents
US8114856B2 (en) 2005-12-02 2012-02-14 Isis Pharmaceuticals, Inc. Antibacterial 4,5-substituted aminoglycoside analogs having multiple substituents
US8569264B2 (en) 2005-12-02 2013-10-29 Isis Pharmaceuticals, Inc. Antibacterial 4,5-substituted aminoglycoside analogs having multiple substituents
US11117915B2 (en) 2007-11-21 2021-09-14 Cipla USA, Inc. Antibacterial aminoglycoside analogs
US8383596B2 (en) 2007-11-21 2013-02-26 Achaogen, Inc. Antibacterial aminoglycoside analogs
US9688711B2 (en) 2007-11-21 2017-06-27 Achaogen, Inc. Antibacterial aminoglycoside analogs
US9266919B2 (en) 2007-11-21 2016-02-23 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8377896B2 (en) 2008-09-10 2013-02-19 Isis Pharmaceuticals, Inc Antibacterial 4,6-substituted 6′, 6″ and 1 modified aminoglycoside analogs
US8399419B2 (en) 2008-09-10 2013-03-19 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8742078B2 (en) 2008-09-10 2014-06-03 Isis Pharmaceuticals, Inc. Antibacterial 4,6-substituted 6′, 6″ and 1 modified aminoglycoside analogs
US8367625B2 (en) 2008-10-09 2013-02-05 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8372813B2 (en) 2008-10-09 2013-02-12 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8524675B2 (en) 2009-05-15 2013-09-03 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8524689B2 (en) 2009-05-15 2013-09-03 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8653042B2 (en) 2009-05-15 2014-02-18 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8658606B2 (en) 2009-05-15 2014-02-25 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8492354B2 (en) 2009-05-15 2013-07-23 Achaogen, Inc. Antibacterial aminoglycoside analogs
USRE47741E1 (en) 2009-05-15 2019-11-26 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8481502B2 (en) 2009-10-09 2013-07-09 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8653041B2 (en) 2010-11-17 2014-02-18 Achaogen, Inc. Antibacterial aminoglycoside analogs
US8318685B2 (en) 2010-11-17 2012-11-27 Achaogen, Inc. Antibacterial aminoglycoside analogs
WO2014166139A1 (en) * 2013-04-11 2014-10-16 北京化工大学 Method for synthesizing dibekacin and arbekacin
CN103204887A (en) * 2013-04-11 2013-07-17 北京化工大学 Method for synthesizing dibekacin and arbekacin
JP2016515589A (en) * 2013-04-11 2016-05-30 ベイジン ユニバーシティ オブ ケミカル テクノロジー Synthesis method of dibekacin and arbekacin

Also Published As

Publication number Publication date
US8399419B2 (en) 2013-03-19
WO2010030704A3 (en) 2010-05-20
US20110275586A1 (en) 2011-11-10

Similar Documents

Publication Publication Date Title
USRE47741E1 (en) Antibacterial aminoglycoside analogs
US8399419B2 (en) Antibacterial aminoglycoside analogs
US8481502B2 (en) Antibacterial aminoglycoside analogs
US8658606B2 (en) Antibacterial aminoglycoside analogs
US8367625B2 (en) Antibacterial aminoglycoside analogs
US8524675B2 (en) Antibacterial aminoglycoside analogs
US8653042B2 (en) Antibacterial aminoglycoside analogs
US8372813B2 (en) Antibacterial aminoglycoside analogs
US8524689B2 (en) Antibacterial aminoglycoside analogs
US20120283209A1 (en) Antibacterial aminoglycoside analogs
US20120283208A1 (en) Antibacterial aminoglycoside analogs
WO2011044498A9 (en) Antibacterial aminoglycoside analogs
EP2217610A1 (en) Antibacterial aminoglycoside analogs
WO2012067978A1 (en) Antibacterial amiinoglycoside analogs

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09792383

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09792383

Country of ref document: EP

Kind code of ref document: A2